ORNL
&EPA
Oak Ridge
National
Laboratory
                                     ORNL/BS-197
United States
Environmental Protection
Agency
Office of Health and
Environmental Assessment
Washington, DC 20460
EPA-600/9-82-001
February 1982
          Research and Development
          Assessment of Risks to
          Human Reproduction and to
          Development of the Human
          Conceptus from Exposure to
          Environmental Substances
          Proceedings of U.S. Environmental Protection Agency-
          Sponsored Conferences:
          October 1-3, 198O, Atlanta, Georgia, and
          December 7-10, 1980, St. Louis, Missouri

-------
     Printed in the United States of America. Available from
             National Technical Information Service
                 U.S. Department of Commerce
        5285 Port  Royal Road, Springfield, Virginia 22161
     NTIS price codes—Printed Copy: A09 Microfiche  A01
This report was prepared as an account of work sponsored by an agency of the
United StatesGovernment, Neither the U nited States Government nor any agency
thereof, nor any of their employees, makes any warranty, express or implied, or
assumes any legal liability or responsibility for the accuracy, completeness, or
usefulness of any information, apparatus, product, or process disclosed, or
represents that its use would not infringe privately owned rights. Reference herein
to any specific commercial product, process, or service by trade name, trademark,
manufacturer, or otherwise, does not necessarily constitute or imply its
endorsement, recommendation, or favoring by the United States Government or
any agency thereof. The views and opinions of authors expressed herein do not
necessarily stale or reflect those of the United StatesGovernment or any agency
thereof.

-------
                                                ORNLyEIS-197
                                             EPA-600/9-82-001
                  Contract No. W-7405-eng-26
 ASSESSMENT OF RISKS TO HUMAN REPRODUCTION
AND TO DEVELOPMENT OF THE HUMAN CONCEPTUS
FROM EXPOSURE TO ENVIRONMENTAL SUBSTANCES
    Proceedings of U.S. Environmental Protection Agency-
                  Sponsored Conferences:

            October 1-3, 1980, Atlanta, Georgia,
                           and
          December 7-10, 1980, St. Louis, Missouri

                      Project Officers:
                  Wayne M. Galbraith, Ph.D.
                     Peter Voytek, Ph.D.
              Office of Research and Development
              U.S. Environmental Protection Agency
                   Washington, D.C. 20460
                           and

                   Michael G. Ryon, M.S.
              Chemical Effects Information Center
                  Information Center Complex
                     Information Division
                 Oak Ridge National Laboratory
                  Oak Ridge, Tennessee 37830

     Work sponsored by the U.S. Environmental Protection Agency,
          Washington, D.C., under Interagency Agreements
              No. 80-D-X1011 and No. 81-D-X0453

                   Published: February 1982
            OAK RIDGE NATIONAL LABORATORY
                 Oak Ridge, Tennessee 37830
                        operated by
               UNION CARBIDE CORPORATION
                          for the
                 DEPARTMENT OF ENERGY

-------

-------
                               IX
                               1
                          CONTENTS

Tables   	
Acknowledgments    	
CHAPTER 1
Introduction    	
CHAPTER 2
Female Reproduction   	    3
  General Reproductive Toxicity Screen	    3
  Qualitative Reproductive Toxicity Tests    	    5
    Estrogen agonist-antagonist    	    5
    Androgen agonist-antagonist	    5
    Nonsteroidal toxicant screening tests     	    6
    Computerized integrated data base    	    6
  Quantitative Reproductive Toxicity Tests     	    6
  Risk Assessment    	    7
  Research Needed    	    9
    Qualitative reproductive toxicity tests    	    9
    Quantitative reproductive toxicity tests     	10
    Specific recommendations	10
    Extrapolation of animal data to humans     	11
References   	                 12
Appendix    	13
Details of Test Protocols and Glossary of Terms for
  Female Risk Assessments	13
Description and Discussion of Tests Useful in Assessing
    Risk to the Female Reproductive System	13
  Qualitative Reproductive Toxicity Tests    	13
    Estrogen agonist-antagonist    	13
    Androgen agonist-antagonist	14
    Nonsteroidal toxicant screening tests     	14
  Quantitative Reproductive Toxicity Tests     	18
    Estrogen agonist-antagonist    	18
    Androgen agonist-antagonist	19
  Hypothalamic-Pituitary Function Tests     	21
    Assay of agents that stimulate the release of
     gonadotropins from cells of the anterior
     pituitary gland    	21
in

-------
   Assays of agents that inhibit the release of
    gonadotropins from cells of the anterior
    pituitary gland    	22
   Assay of an agent that inhibits the release of
    prolactin from  cells of the anterior pituitary
    gland	  23
   Assay of an agent that inhibits the release of
    prolactin from pituitary cells  	23
   Assay of the activity of an agent that alters the
     secretion of dopamine by hypothalamic
     neurons	24
   Assay of the activity of an agent that alters the
     secretion of norepinephrine by hypothalamic
     neurons   	24
   Assay of the activity of an agent that alters the
     secretion of GnRH   .	•  •  24
   Assay of the activity of an agent that alters the
     secretion of hypothalamic opioid peptides	25
   Blood flow of the hypothalamic-hypophysial system     .  .   25
   Sexual behavior  tests	25
  Ovarian Toxicity    	27
   Oocyte and follicle toxicity    	27
   Inhibition of steroidogenesis	28
References   	34
Glossary of Terms Used in Female Reproduction    	37
CHAPTERS
Considerations in Evaluating Risk to Male
  Reproduction    	41
Introduction    	41
  Aspects of the Problem    	42
  Selection of an Animal Model   	43
  Tests for Evaluating Reproductive Damage    	46
  Evaluation of Reproductive Damage in Exposed or
     Potentially Exposed Men    	52
    General	52
    Surveillance studies   	52
    Study of men with known toxic exposure	53
    Additional comment on human testing procedures    .  . .54
    Assessment of risk to men	55
  Protocols for Testing Compounds with Animal Models    . .  57
    Test 1 — initial screen     	57
    Test 2 — dose response curve	57
    Test 3 — recovery study     	59
                                iv

-------
  Research Needed   	60
References   	'.	63
Appendix    	69
Details of Test Protocols and Glossary of Terms for
   Male Risk Assessment    	69
  Description and Discussion of Tests Useful in
   Animal Models or Man    	69
  Body weight	69
  Testicular Characteristics   	69
   Testis size in situ    	69
   Testis weight	70
   Spermatid reserves	70
   Histopathological analysis of testes    	71
   Counts of preleptotene or leptotene
     spermatocytes    	72
  Epididymal Characteristics   	72
   Weight of distal half of epididymis    	72
   Number of sperm in the  distal half of
     epididymis   	72
   Motility of sperm from the distal end     	72
   Gross morphology of spermatozoa from the
     distal end	73
   Detailed morphology of  spermatozoa from the
     distal end	  73
  Accessory Sex Gland Characteristics    	73
  Seminal Analysis    	73
   General aspects of seminal analysis    	73
   Volume	75
   Seminal plasma constituents	75
   Spermatozoa! concentration	76
   Total sperm per ejaculate   	76
   Sperm motility    	77
   Spermatozoal morphology     	78
   Ejaculated sperm as an in vitro test system    	79
  Assessment of Male Reproductive Toxicity Using
     Endocrinological Methods   	79
   .General	     79
   Hormone assay and application	80
  Examination of Known Toxic Exposures	82
   Humans	82
   Animal models    	83

-------
 Fertility Testing    	.84
   Tests available    	84
   Usefulness	85
   Sensitivity	85
   Specificity	86
 Sperm Nucleus Integrity    	86
   Quinacrine staining for Y-chromosome
     aneuploidy	86
   Spermatozoal morphology    	87
   Karyotyping of human spermatozoa by the
     denuded-hamster-egg technique   	87
 Dose Response	87
References    	88
Glossary of Terms Used in Male Reproduction    	93
CHAPTER 4
Current Status of, and Considerations for, Estimation
   of Risk to the Human Conceptus from
   Environmental Chemicals   	99
 Definition and Scope     	99
 Impact of Developmental Abnormalities on Humans    ...   99
 Causes of Congenital Malformations	100
 Qualitative Evaluation of Risk Potential    	100
   Interspecies comparisons   	100
   Dosing and mode of administration   	101
   Placenta! transfer   	102
   Pharmacokinetics and metabolism	103
   Mechanisms of action    	103
 Animal Studies	104
   Standard teratogenicity testing	104
   Functional teratogenicity testing   	106
  Short-Term Testing Procedures     	107
   Prioritizing of chemicals for in-depth study    	107
   Characteristics of short-term assays   	108
   Potential short-term systems	109
  Quantitative Risk Assessment	110
  Priorities for Future Research in Teratology    	112
References   	113
CHAPTERS
Other Considerations: Epidemiology, Pharmacokinetics,
    and Sexual Behavior   	117
  Epidemiology: Methods and Limitations   	117
    Hypothesis generating studies    	117
                               VI

-------
    Analytic studies for formally testing hypotheses
      and quantifying risks   	118
    Limitations	118
    Possible data sources and useful approaches   	120
  Pharmacokinetics  .  „	  .  121
  Sexual Behavior   	123
    Introduction	123
    Qualitative evaluation of risk potential	128
    Animal studies   	131
    Assessment of human sexual behavior:
      surveillance and epidemiological
      studies    	136
    Priority areas for future research    	139
References   	141
Steering Committee	145
Participants	'         147
Reviewers    	151
Index    	153
                               VII

-------

-------
                           TABLES

 1. Reproductive Processes Potentially Susceptible
      to Reproductive Toxicants     	    8
 2. Estrogen Agonist Screen    	14
 3. Time Table for Intergenerational Protocol to Evaluate
      Putative Toxicant Effects on Reproduction in Sexually
      Mature Animals	15
 4. Compounds Tested for Oocyte/Follicle Toxicity in
      the Murine Assay    	29
 5. Agents That Inhibit Steroidogenesis	31
 6. Features of Ovarian Cell Preparations, In Vitro,
      Potentially Useful in Xenobiotic Inhibition of
      Steroidogenesis   	33
 7. Criteria for Evaluation of Male Reproduction in
      Favored Animal Models and Man   	44
 8. Te,sts Considered Useful for Screening Toxic
      Compounds    	47
 9. Reasons for Rejection of Potential Evaluation
      Tests Considered by Male Reproductive
      Subgroup    	49
10. Approximate Variation Between Animals for
      Suggested Test Criteria (CV) Coefficient of
      Variation (%)	50
11. Chronology of Conduct for Test with Animal
      Models   	58
12. Some In Vitro Short-Term Systems Currently
      in Various Stages of Development	110
                               IX

-------

-------
                    ACKNOWLEDGMENTS

    This document  resulted from  discussions at two  conference
meetings sponsored  by the  U.S. Environmental Protection Agency
(EPA).  The first meeting took place in Atlanta, Georgia, October
1-3, 1980, and the  second, in St. Louis, Missouri, December 7-10,
1980. The participants in this project are listed at the back of this
document; their interest, scientific knowledge, and contributions of
personal and  professional  time  are  largely  responsible for  the
production  of  this report. The  steering committee and the group
chairmen  contributed  additionally  by selecting participants, orga-
nizing  subject  agenda, and refining  reports produced  during the
conferences. In particular, Drs. Richard, Hoar and Marshall Johnson
played major roles throughout the duration of the project. The final
step involved external  reviews, and the contributors to this process
are also listed in the back of this report.
    In addition to the  input from the scientific community,  efforts
of tne technical staff from Oak Ridge National Laboratory played a
large  role   in  the  success of  the  project.   Joy Simmons   and
Norma Callaham handled arrangements for hotel accommodations
and equipment rental.  Debra Ballard,  Evelyn Daniel,  PatHartman,
Robert Ross, and John  Smith provided word processing and logistical
support for the participants during the conferences. Members of the
Technical    Publications   Department,   especially   Pat Hartman
and Donna Stokes,  were responsible  for typing,  and John Getsi,
for editing tne drafts into finished form.
    The U.S. EPA's  Offices of Research and Development  and of
Pesticides   and  Toxic   Substances  and  the  Oak  Ridge National
Laboratory gratefully acknowledge  the efforts of all those involved
and thank tnem for assisting in this project.

              Dr. Wayne Galbraith,  U.S. EPA Co-Project Officer
              Dr. Peter Voytek, U.S. EPA Co-Project  Officer
              Michael  Ryon, ORNL Conference Coordinator
                               XI

-------

-------
                         CHAPTER 1
                      INTRODUCTION
    The  U.S. Environmental Protection Agency  (EPA)  has the
legislative mandate to consider regulatory alternatives for chemicals
that are causing or can cause a health hazard to man. Because the
reproductive system contains some of the more sensitive targets of
potentially  hazardous agents whose impact on human populations
may  be immediate,  tpxicity to  the reproductive  system  and the
conceptus is of emerging scientific and social interest. As a result of
this interest, the  Offices  of Health Research and of Health and
Environmental Assessment within  the  Office of Research and De-
velopment  sponsored a conference to produce a technical document
on the current status of risk assessment methodologies  for terato-
genic and  other reproductive effects. The  conference brought to-
gether scientists knowledgeable in reproductive  biology and tera-
tology to discuss  techniques and  concepts pertinent to  developing
risk assessment methodologies.
    Conference participants were selected based on their expertise in
the various  disciplines of reproductive biology, statistics,  pharmaco-
kinetics, endocrinology, epidemiology,  and sexual  behavioral toxi-
cology. Draft copies of the report were sent to numerous scientists in
academia and the private sector for peer review, and their comments
were  used  by the  members of the conference to modify the final
document.
    The document is  divided into  three main subject areas: assess-
ment of toxicity to female reproduction, assessment of toxicity to
male  reproduction, and assessment of toxicity to the conceptus.
There  are  three supplemental parts: pharmacokinetics  and epide-
miologic considerations, which  are common  to  all toxicological
assessments, and a  special section on the behavioral aspects of sexual
development.
    The  specific areas addressed  in  this report  are the potential
adverse effects on  the female and male reproductive systems as well
as adverse  effects  on the developing conceptus. A broad range  of
problems and  effects  are  discussed,  including infertility,  early
resorption  of the conceptus, and possible  behavioral disorders

-------
produced by subtle changes in the biochemical environment of the
fetus.
    The report also provides suggestions for improvement in standard
toxicological protocols for evaluation of reproductive risks, identifies
new concepts and procedures that  can be immediately applicable,
and designates those that need further expansion and development
through research. Included is a discussion on the predictive ability of
the tests in estimating risk.
    The information in this document will be of value not  only to
scientists  conducting experiments on the effects of chemical agents
on the reproductive system, but also to those that need to assess the
results from such  studies.  Thus many tests discussed herein may
currently be inappropriate, economically or technically,  for regula-
tory use, but  are  included to  provide necessary and useful back-
ground information for evaluating data.
    In assessing human risks  from  exposure to potentially toxic
chemicals, many considerations should be addressed, such as  severity
and reversibility/irreversibility of the effect, existence of threshold or
nonthreshold levels, dose-response relationships, sensitivity of the
toxicological response  evaluated, and predictive ability of animal
studies to determine the risk to humans. Attempts have been  made in
this document to address these considerations.

-------
                         CHAPTER 2
                FEMALE REPRODUCTION
    Risk assessment for toxicants that alter reproduction in females
involves two separate but equally critical tasks. These are assessment
of reproductive parameters in laboratory animals to identify com-
pounds that are prospective reproductive toxicants  and continuous
epidemiologic surveillance of normal human reproductive character-
istics to  identify their  prevalence, trends,  and geographical  dif-
ferences and their potential modification by environmental events.
An approach to the evaluation of epidemiologic data is provided in
Chapter 5.
    The  problem of  risk  assessment  has  been  approached by
proposing an animal screening system for qualitative and quantitative
analysis  of reproductive toxicants. This system is coupled to an
integrated  data base that serves as  a mechanism for the analysis of
structure-function relationships  of  potential toxins. These  testing
systems form a comprehensive screening scheme that should serve to
detect  reproductive  toxins  and provide  a foundation for  risk
assessment. In addition,  such a system will serve as a repository of
information into  which continued input should expand our under-
standing of risk assessment and reproductive toxicology.
              General Reproductive Toxicity Screen

    We propose that the stepwise scheme shown in Fig. 1 be followed
in an attempt to identify compounds presently in the environment
for which  there is epidemiologic evidence of adverse reproductive
effect and to identify new compounds that may be disseminated into
the environment. At the first level, a compound should be tested by
tne laboratory procedures described below. The standard acute,
subacute,  and  subchronic  toxicological  testing  protocols  do not
incorporate procedures for detection of reproductive effects, and
therefore the  following screening procedures  were  specifically de-
signed for this purpose. If the result of any screening test is positive,
the compound must be evaluated by the quantitative risk assessment
procedures. If the screening  tests  are  all negative,  the  compound

-------
                                            ORNL-DWG 82-8409
                 Qualitative
                toxicological
                   screen
                                   Computerize
                                     integrated
                                     data base
    Qualified
     release
Quantitative
toxicologicai
   screen
                                Release without
                                    further
                                  qualification
                Risk assessment
                   decision
      Figure 1 - Reproductive toxicity assessment prospective evaluation.
must then be compared by the computerized integrated data base for
structural relationships and/or similarities in the probable pharmaco-
kinetics  witn other compounds known to affect the reproductive
system. An examination of the potential degradation products of the
compound,  using  a  computer  model analysis of its  chemical
structure, might also prove useful. If the compound is found to have
a structural or functional similarity to known active agents, it must
undergo the  quantitative risk assessment procedures.  If  no such
affinity is found (and the qualitative screen is entirely negative), the
compound  can be released  into the environment without further
testing.

-------
                                 5

    If a  compound that  enters the  quantitative risk assessment
 procedures is found to be without activity ("false positive" in screens
 or in search for structural affinity), it can be given qualified approval.
 Tnat is, its use must be restricted and accompanied by appropriately
 directed epiaemiologic surveillance.
    If a  compound that  enters the  quantitative risk assessment
 procedure is positive,  the  risk for humans  should be estimated,
 insofar as possible. This information  must  be  weighed and  the
 decision made about whether the compound can be released at all
 and if so, with what restrictions.
              Qualitative Reproductive Toxicity Tests

Estrogen agonist—antagonist

    Estrogens mediate, integrate,  and modulate interaction of the
hypothalamic-hypophyseal-gonadal axis and, as such, are important
hormones in the  control of reproduction. Exposure to  exogenous
estrogens is known  to have  deleterious effects  on  reproductive
potential  (1).  For predicting  the estrogenicity of environmental
chemicals,  a series of simple  screening tests  are  proposed.  These
include (a)  time  of vaginal opening in the neonatal rat, (b) uterine
epithelial cellular hypertrophy,  and  (c)  estrogen-receptor-binding
analysis. These tests have been chosen because of their  sensitivity to
estrogenlike substances and the close correlations that exist between
these estrogenic  responses and subsequent  abnormal  reproductive
capacity. For details of the test procedures, see the Appendix to this
chapter.
    The tests cited above can  be used to detect estrogen toxicants;
however,  they could  also be used to  classify  estrogen agonist-
antagonist.  Generally,  a decreased  response  in  the  tests would
indicate an antagonistic effect, whereas an increased response would
indicate an agonistic  effect.  Such a classification scheme, which
would require extension and expansion of the tests cited, could form
the basis for  a structure-function  data bank for the prediction of
estrogenic toxicity.

Androgen agonist—antagonist

    Androgenic substances are  known to cause infertility in female
animals, and  their effects  on the human  fetus are well known.
Exposure to androgens during pregnancy causes masculinization of
female fetuses and various physiological and  behavioral problems in

-------
the adult. Androgenic compounds can be assessed by their capacity
to stimulate weight increases  in  the ventral prostate and seminal
vesicles of the intact immature male  rat or mouse. This assay is
acceptable and practical for predicting androgenic effects (agonistic
and/or antagonistic) in humans.
    It  is  important  to determine  whether a potential toxicant
influences development or reproductive capabilities. For this pur-
pose, determining responses of newborn rats following exposure to
suitable  doses of the potential toxicant provides  a multifactorial
assay.  Similarly,  screening tests (e.g.,  testosterone  blood levels or
accessory  sex gland  weight)   for  androgen  antagonists  are also
available. More sensitive tests  for androgen agonists/antagonists are
described in the quantitative section of Female Reproduction and in
the Appendix to this chapter.

Nonsteroidal toxicant screening tests
    The   preceding  tests  will detect  estrogenic  and  androgenic
toxicants. For testing of substances other than these two classes of
compounds, a multigenerational protocol is proposed. This protocol
is designed to evaluate (a) adult female conceptive ability with initial
exposure to  the  agent occurring  near puberty,  (b) the effect  on
pregnancy,  (c) potential  transmission  during lactation,  and (d)
reproductive performance of  the second generation. As  a  standard
approach  to the  testing of potential  reproductive  toxicants, these
tests will  detect  substances  that  interfere with  reproduction at
various  levels  of biological   organization. (For   details see  the
Appendix.)

Computerized integrated data base
    The computerized integrated data base should include all known
structure-function relationships for reproductive toxicants. With such
a data base it would be possible to construct reproductive toxicant
profiles (activity profiles)  that would predict the potential activity of
putative toxicants. Admittedly, such a scheme has shortcomings and
prediction will not  be perfect.  However, if  at some  future date
 sufficient information were available in the data base, it could prove
 to be most useful and time saving.
              Quantitative Reproductive Toxicity Tests

     The screening tests outlined in the previous section are designed
 to  identify compounds that may represent reproductive hazards.

-------
                                7

Once a compound is found to have either a reproductive effect in the
qualitative screen or a  structural  relationship  with known active
agents in  the  computerized-integrated-data-base  screen,  a  more
detailed quantitative evaluation is mandatory. This process will vary
substantially, depending on the type of effects seen in the qualitative
screen or  the  characteristics  of the known toxicant  to  which it
appears similar. These tests for quantitative assessment, which are
presented in  detail in the Appendix to  this chapter, can be used to
determine site or locus of action of these xenobiotics. It is important
to recognize  that  some xenobiotics may act at more than one site
and by more than one mechanism. Ultimately, these specific assays
have the potential to determine risk of  exposures. In this document
an  attempt  is made  to  provide  an  interface  between  female
reproductive biology and toxicology.
                        Risk Assessment

    Assessment of  risks to  the  female reproductive  system from
environmental  sources will have to  involve a  broad  class  of
potentially affected processes, organs, and  structures obtained from
human exposure and relevant laboratory results. The reversibility of
effects needs  to  be considered carefully.  The applicability of the
available information to potential environmental exposure will need
to be considered. The magnitude  of human risk for reproductive
toxicity may be modulated  by such diverse factors as distribution of
the compound in  the environment, patterns  of use  or exposure,
persistence in the biosphere, concentration in  the food chain, and
age-dependent changes in sensitivity.
    Risk assessment will require a knowledge of pertinent factors
related to the reproductive process and of relationships of specialized
laboratory results to  these  factors. If a compound demonstrates a
reproductive effect in any mammalian species, this observation
indicates that some concern about actual human exposure to  the
agent is  justified. Positive results in a number of laboratory tests,
which  by  themselves  may  be  only  suggestive  of  harm,  will  be
important in evaluating potentially detrimental effects.
    Substantial modifications in any  of the  subsystems given in
Table 1  are known to be  serious  and  should be avoided. Future
testing may  indicate  relationships between these subsystems and
other laboratory  testing. Risk assessment  for  female  reproduction
requires  the establishment of assays relevant to these reproductive
processes and  the validation of these assays in identifying substances
actually  toxic to  human reproduction. The assays should be shown

-------
                   TABLE 1 Reproductive Processes Potentially
                      Susceptible to Reproductive Toxicants
                             Nonpregnant
                                                                Pregnant
Vulva/Vagina         Viiilization
                     Adenosis
Cervix               Structural abnormalities
                     Mucus production and/or
                       quality
Uterus               Luminal fluid
                     Structural malformations
                     Dysfunctional bleeding
                     Dyssynergia
                     Deficient pseudodecidual
                       response
Fallopian Tube       Gamete transport fluid

Ovary               Decreased number of oocytes
                     Increased rate of follicular
                       atresia
                     Follicular: steroidogenesis
                               maturation
                               rupture
                               fluid quality
                     Oocyte maturation
                     Luteal function
                     Chronic anovulation
Breast               Supernumerary mammary glands
                     Galactorrhea
                     Nongalactorrheic discharge
                     Gynecomastia
Placenta
Pituitary             Hyperprolactinemia
                     Hypoprolactinemia
                     Altered synthesis and
                       release of trophic
                       hormones
Hypothalamus       Altered synthesis and
                       release of neurotransmitters,
                       neuromodulators, and
                       neurohormones
Liver                Metabolism
                     Binding protein synthesis
Adrenal             Steroidogenesis
Behavior             Sexual behavior
Reproductive        Puberty
  lifespan            Menopause
Incompetence
Untimely parturition
Dysfunctional labor
Uterine blood flow
Gestational trophoblastic
  disease
Deficient decidual response
Zygote transport
Ectopic pregnancy
Luteal function
Lactational transport of
  toxicants
Lactation: composition
  capability
Transplacental transport
  of toxicants
Hydatidiform mole
Enzymatic activities
 Metabolism
 Binding protein synthesis
 Steroidogenesis
 Maternal behavior

-------
to be relevant in the species used, especially any assays adapted from
human assays, and should avoid complications such as those resulting
from diurnal variations  in  hormone level. The  results  from  these
studies, may indicate other areas where regulation will be necessary.
    It is generally  felt  that  when  dose-response relationships are
observed, lowering exposure  will cause less  harm when the  com-
pound is xenobiotic, unless evidence to the contrary exists.  Con-
sideration  could  be given  to  using  safety  factors to  establish
acceptable exposure levels in situations where harm can reasonably
be expected and the exposure cannot reasonably be avoided.
                        Research Needed

Qualitative reproductive toxicity tests
    The scheme for the detection of reproductive toxicity discussed
earlier and diagramed  in  Fig. 1 proposes  further investigation in
several research areas. One such area is  that of structure-function
relationships, which are not well understood at the present time. No
one would have predicted from the structure of kepone that it would
bind  to  the estrogen receptors and stimulate estrogenic responses.
Obviously, much  needs to be learned about  what  constitutes  an
estrogenic  molecule. However, kepone would have been detected as
an  estrogen by the above  tests, and indeed, had more been known
about structure-function relationships, it might have been suspected
before any tests were performed.
    The  establishment  of reproductive-toxicological profiles and
structure-function prediction models  has just  begun. Much  basic
information is required  before  such a system can  be realized.
Therefore, a  strong recommendation  is  that  basic  research  in
reproductive  toxicity  be  supported,  with a major emphasis  on
establishing such models.
    An important component of the qualitative reproductive toxicity
screen is the  computerized integrated data base (see Fig. 1).  With
such  a data base, it should be possible to predict the  potential
toxicity  of putative toxicants. That such a predictive scheme has its
faults  is well recognized; however,  further efforts  to realize the
potential of such a system should not  be discouraged on account of
these. In theory, when sufficient  information is available concerning
structure-function relationships  of toxicants,  such a prediction
scheme may decrease.the need for extensive animal testing. For this
reason it is recommended that further attempts to establish and

-------
                                10

validate such  a data base be made and also that arrangements be
made to update continuously such a facility, with the ultimate intent
of perfecting predictive potential.

Quantitative reproductive toxicity tests
    The recommendations concerning the qualitative tests also apply
to the proposed quantitative tests. The information gained from the
quantitative tests extend and interplay with  the results obtained
from  the  qualitative tests.  Therefore,  it  is  recommended that
information gained  from the quantitative tests be integrated with
that obtained from  the  qualitative tests to permit an even greater
understanding of structure-function relationships.


Specific recommendations
    1. The  relationship between cellular receptors for  toxicants and
their mechanism of action  should  be  explored  further.  Such
information can  be  fed directly into  a  structure-function data bank
such as the computerized integrated data base.
    2. More work is obviously needed  regarding masculinization of
the  female, an important problem in reproductive toxicity evalua-
tions. Few data are  available on dose responses of these effects, and
fewer  data exist regarding inhibition  (antagonism)  of the alteration.
Further, extrapolation of  these  data  to humans is  not possible,
because subhuman primates and humans do not sustain substantial
defects of ovulation, whereas sexual behavior is altered. Information
presently  available  is insufficient  for determining  whether  this
discrepancy  is  due to  the  fetal age  at  which treatment  was
administered or to actual differences in sensitivities.
    3. In vitro model systems are needed (in many areas)  for the
assessment of reproductive toxicants. For example, model systems
for the secretion of gonadotropins by the pituitary cells can be used
to  study  toxicants that influence this process.  Currently, almost
nothing is known about such model  systems, and their value  for
predicting toxicity is potentially great.
    Another important in vitro model system in need of development
is that of inhibition of  steroidogenesis. Although this system has
been well characterized for many inhibitors (see the Appendix to this
chapter), it has not  been exploited for its potential as a test system
for  toxicants. Continued work and support will be needed to develop
these model systems and to relate the  information obtained to that
gathered from in vivo studies.

-------
    4. Because little is known about  the  effects of  neuroactive
 substances   such  as  dopamine  and   norephinephrine  on  the
 hypothalamic-hypophysial complex and about the effects of toxi-
 cants  on this system, research support  should be allocated to this
 important field,  both for the development of new methods and for
 studies on the mechanism of action.
    5. Support is recommended for  development  of models  and
 research on basic mechanisms in behavioral  toxicology, an area in
 which many unknowns exist regarding reproductive toxicity (see the
 Appendix).
    6. Much work  is  needed in  oocyte toxicity,  an  obviously
 important area of concern  in  which there are incompletely under-
 stood age, strain, and species differences in sensitivity of ovotoxicity.
 For example, preovulatpry and growing follicles are most sensitive to
 toxicity in  humans, whereas resting  follicles are most  sensitive in
 mice.  Similarly,  significant differences in   sensitivity  to  oocyte
 destruction exist  between mice and rats. However, evidence from
 studies exploring  the effects of antitumor agents on humans  and
 experimental animals suggests that a compound demonstrated to be
 ovotoxic in rodents will also be ovotoxic in humans.

 Extrapolation of animal data to humans

    The primary goal of risk assessment for environmental agents is
 directed toward adverse effect(s) (injury) in  human individuals or
 human populations. In most cases data are available only in animal
 model systems; hence it is necessary to extrapolate these findings to
 anticipated  changes in humans. Although extrapolations  may be
 possible, it  should be noted that our current understanding of the
 relationships between hormone exposure and  toxic outcomes is  not
 optimal. The following discussion is  included  to illustrate this point.
    An increased  rate  of vascular  disease in women  taking oral
 contraceptive pills has been reported by several investigators (see Kay
 [2] for review). This has been  generally attributed to the estrogenic
 component  of the pill and at first may seem to represent a source of
 data concerning estrogen levels and  toxic  effects. However, as Kay
(2) points out, the progesterone content of the pill, not the estrogen
content, is  correlated with increased incidence of vascular disease.
Progesterone has  also been shown  to decrease high-density lipo-
protein (HDL) cholesterol, an event associated with increased risk of
arteriosclerosis. Estrogens increase HDL cholesterol and therefore
would be expected  to  decrease  the  incidence of vascular disease.
Obviously, predicting risks based on estrogen levels in women taking
the pill requires further consideration.

-------
                                 12
                           t
    Tliis example points out the need for more research at all levels,
from  biochemical to epidemiological,  and emphasizes the require-
ment for  more data before  meaningful extrapolations can be made
for risk assessment in humans.

                          REFERENCES
1. McLachlan, J.: Estrogens in the environment. Elsevier/North Holland: New
     York; 419 pp., 1980.
2. Kay, C. R.: The happiness pill? J. R. Coll. Gen. Pract. 30: 8-19, 1980.

-------
                         APPENDIX
          DETAILS OF TEST PROTOCOLS AND
               GLOSSARY OF TERMS FOR
              FEMALE RISK ASSESSMENTS

       I.  DESCRIPTION AND DISCUSSION OF TESTS
            USEFUL IN ASSESSING RISK TO THE
             FEMALE REPRODUCTIVE SYSTEM
             Qualitative Reproductive Toxicity Tests

Estrogen agonist—antagonist

    Time of vaginal opening in the neonatal rat. Rats are injected on
days 1,3, and 5 of postnatal life, and the time of vaginal opening is
noted.  Estrogen  agonists such as  diethylstilbestrol (DBS), clomi-
phene, and tamoxifen are known to cause early maturation of vaginal
development (1—3),  and this test serves as a sensitive indication of
such activity. The general protocol for this test for estrogen agonist is
shown in Table 2.
    Uterine epithelial cellular hypertrophy. Neonatal rats are treated
as described  in Table  2, and the  uteri are  taken on  day  7 for
histological  examination. Epithelial cell  growth  is an  excellent
indicator  of  estrogenicity and  will detect compounds,  such as
clomiphene, which exhibit differential cell stimulation (4). Kepone,
DES, dichlorodiphenyltrichloroethane (DDT), and  zearlenone have
been shown  to  be either active in this test or very  likely  active
because of their known ability to stimulate uterine growth (4, 5).
    The above tests, requiring a minimal number of  animals, are
simple  and  reliable.  These tests   are  used routinely  in  many
laboratories and are quite sensitive to estrogenic compounds (/zg/kg).
    Estrogen-receptor-binding analysis.  Uteri obtained  from 7-day-
old rats which have been treated as described above are examined for
nuclear binding of the estrogen receptor by the nuclear exchange
assay (4). In  the same tissues  the quantity of cytoplasmic estrogen
receptor can  also be determined. This  test  gives a measure of the
                              13

-------
                                 14
                     TABLE 2 Estrogen Agonist Screen
                Age
               (days)
Treatment of
 female rats
                 0         Birth
                 4         Beginning of daily dosing for 4 days
                 7         Sacrifice
                           Assays:
                           1 — uterine weight
                           2 — endometrial histology
                           3 — estrogen receptor assays
                           4 - vaginal opening0

                 "Vaginal opening may occur by day 7; however,
                a longer time interval after birth may be required
                (up to 20 days).
ability of a toxicant to bind to estrogen receptor in vivo and to cause
nuclear  accumulation  of the  receptor-ligand  complex.  Classical
estrogens such as estradiol  and DBS  are  known  to  perform  this
function, which  is  presumed  to be  an  obligatory step  in  the
mechanism  of action  of estrogens. Kepone, DDT, and zearlenone
bind  to the  estrogen receptors,  cause nuclear accumulation,  and
stimulate uterine growth (3, 5 6).  Therefore, these compounds are
likely to be active in the other tests for estrogenicity and will make
excellent reference compounds for testing the model.

Aiidrogen agonist—antagonist
    These tests are standard and require no further explanation.

Nonsteroidal toxicant screening tests
    This protocol is  designed to  evaluate: (a) ability of the adult
female to  conceive with the  initial  exposure to the agent  near
puberty (P-generation, Table 3); (b) the effect on pregnancy  (live
birth index of Fj, F\, F2, F2 generations); (c) potential transmission
during lactation (survival  index of Fj  and F2  generations); and (d)
reproductive performance of the second generation (live birth index
 of Fj  and F2 generations). Part of the P,  generation (PJ) is mated
 again at the time of postpartum estrus, because at that time mating
 behavior,  ovulation,  implantation, and fetal  resorption are  more
 sensitive to environmental disruption than they are during mating at
 a cycling estrus.

-------
15






o
W
c
s
o
0>
1 „
(0 g
i'l
w 3
o £j
s i
O 3
•a W
2 £
-2 'S
£3 -2

l|
•2 a.
.0 C
«w o

1
1
uT

tf
»> •« ri
.S E — fc
Iffe l^
**« •*"" -"K C T3
O *-> '~* o C
13 H M " °
"^ ^~* sn w So
HI I i
^ C X> G 43
III II
tiT uT ciT oT uT


"O
ZJ
.s
'c ^

^o rt
I 3 1 1
QJ ^ •*-• "^
op ^3 M a, tL<
c .E S 2 » S
i^ w aj ca O rt
1 1 1 1 (JI 1
B. CL, CL, CL, CL, ft.



Of*^ \om»oo\O<^
^^T T7T7TT













.i
| oil
ll

-------
                                16

   The age at which the animals are mated is a major procedural
factor  that can Wluence the fertility test results in the F1 and F2
generations. Toxicants,  particularly  those that  possess  steroidal
activity,  will  diminish  the success of pregnancy and  number  of
offspring of older females but not those of younger females. Thus,
while testing at earlier ages is more economical, it might yield  false
negative results. Therefore, a portion  of the Fj  generation should be
examined for ovarian cyclicity and  fertility at  approximately six
months of age (live birth index of F^ generation).
    Experimental, vehicle  control, and positive control (use  of a
known toxicant to verify the system) groups should be utilized with
at least 20 animals per  group. Selecting the agent to be used as a
positive control will  be arbitrary,  and species  or strain differences
may complicate the choice. Despite these drawbacks, inclusion of a
positive control that most appropriately parallels the test compound
would seem mandatory for validation of the test system. The entire
protocol need not be completed  if adverse effects are demonstrated
early in the protocol (i.e., live birth index of the Fj generation).
    The maximum tolerated dose should  be used. Other dose levels
may be included if dose response information is needed.  Route of
administration should be in food  or water  to avoid handling pregnant
and lactating females, which may result in stress independent of that
potentially caused by the agent  being tested.  This may complicate
quantification of the ingested dose but ensures continuous dosing of
the  Fj generation during weaning. Other routes of exposure  (e.g.,
gavage or parenteral administration) may be used if the test protocol
can be modified to avoid any interfering stress. Dosing begins at six
weeks of age of the Pt  generation and continues until the end of the
protocol. Body weights  should be recorded weekly for all animals in
the P! and F: generations as well  as pup weights in the Fj, FJ, F2,
and F2 offspring.
    P! females are mated with untreated males of proven fertility at
ten weeks of  age in a one-to-one sex ratio.  Successful  mating is
determined by  finding a copulation  plug  and  presence of sperm in
the vaginal smear. These same females are then mated  again at the
time of postpartum  estrus,  8-10  hours  after  giving birth. Twenty
females from the Fl generation (the offspring resulting from the first
mating), are randomly selected  and  mated with untreated males of
proven fertility  at ten  weeks of age. The offspring of postpartum
mating (FJ) need only be counted and weighed  at birth.
    The selection of the species of the test animal to be used in the
 toxicant  screening procedures will be determined  by  several con-
 siderations including cost, time,  and ability to assess related human

-------
                                 17

 reproductive  processes. The rhesus  monkey  or other subhuman
 primates, being a more comparable reproductive species, would be
 the animals of choice, but their cost as well as other considerations
 would prohibit their use in screening procedures. Laboratory rodents
 are economically feasible, but the relevance of the outcome of the
 screening to the human could be questioned. On the basis of current
 information, different species and strains will have to be selected for
 evaluating different  components of the human reproductive system.
 Each choice would carry with it a risk of obtaining false positive and
 false negative  data with regard to the relevance to the human female.
 For example,  on the basis of contemporary results, the rat would be
 less satisfactory  than  the guinea pig for  assessing the effects  of
 potential toxicants  on the development  or the  integrity of cyclic
 gonadotropic function.
    Several neural and physiological interventions that curtail estrous
 cycling in the rat do not occur in the rhesus monkey and guinea pig.
 In  addition at least some perinatal steroid manipulations that render
 the rat permanently anovulatory apparently do not  interfere  with
 menstrual  cycles in the rhesus monkey. Thus, it is likely that many
 substances found to disrupt spontaneous ovulation in the rat will not
 do  so in the human, and false positive assessments may result.
    A false negative may occur if the rat is the  only species used  to
 assess the reproductive consequences of a compound. For example,
 the ovarian cycle of the rat does not have a spontaneous luteal phase
 as does the human cycle. Therefore, compounds that might interfere
 with the function of the corpora lutea cannot be detected in the rat.
 Under  these  circumstances  another  species with  a comparable
 reproductive process, such as the guinea pig, should be considered for
 addition to the screening procedure. .
    Indexes should  be calculated for mating, fecundity, female
 fertility, and parturition as noted below.

 mating index = numbe? of copulations (one counted/estrous cycle)
                       number of estrous cycles required
                                                         x  100
fecundity index =
                 numer
                 number of copulations

fertility index = numb^r of fi^f conceiving
                number of females exposed

incidence of parturition = num^er °£ Parturitions
                        number of pregnancies

-------
                                18
   Numbers of viable,   stillborn,  and cannibalized progeny  are
recorded for each litter,  the survivors on  days 1,  4,  12,  and 21
postpartum noted, and litters reduced to ten pups on the fourth day
of lactation for standardization. Gestational length and sex ratio are
also monitored.

    ,.,,.,    number of viable pups born   1fin
live birth index = total number of pups born x  1 °°

              .  ,   number of pups viable at location day 1 or 4
1- or 4-day survival =	number of pups born	
  index
       _,  ,       .  , _ number of pups viable at lactation day 12 or 21
12- or 21-day survival -   number of pups retained at lactation day 4
  index

             Quantitative Reproductive Toxicity Tests

Estrogen agonist—antagonist
    The screening test listed  previously under qualitative assessment
can be used to  establish  dose-response relationships between estro-
gens  and suspected estrogenic toxicants. The following discussion
represents an expansion of the qualitative tests.
    Neonatal exposure to various dose levels of estrogenic toxicants.
These assays will result in a dose-response relationship  for time of
vaginal opening, ovarian  degeneration and oocyte loss, and stimula-
tion of epithelial cell height  in the uterus. These end points are easy
to assess, are reproducible, and are quite sensitive (jug quantities of
DBS, Kepone, and clomiphene are easily detected) (1-3). This is not
to say that these tests have been utilized to examine a large class of
compounds; however,  one  of the recommendations is that such
compounds be  studied in detail. At the  present time  all known
estrogens are active in these assays, and hence we  can  expect  that
they  will be good  predictors of estrogenic potency. Likewise, such
assays should identify compounds that may interfere with reproduc-
tive  processes.  It  may be  possible to extrapolate these  data on
relative potency to known effects of various doses of estrogens in the
human,  since it is  well  established  that  estrogenic responses in
rodents  and humans show many similarities (7—9). To  this  end
 compounds such as ethynylestradiol,  DBS, and estradiol should be
 used as standards.
    Estrogen receptor  analysis in vivo  and in vitro. An extremely
 sensitive  (picogram-nanogram  range)  and reproducible  method for

-------
                                 19

 assessing relative  estrogenicity  involves  the use  of toxicants to
 compete with labeled estradiol in binding to uterine cytoplasmic
 estrogen receptors (3-6). This test involves the addition of various
 concentrations  of the toxicant  to uterine  cytosol fractions in the
 presence of labeled estradiol. If the toxicant is estrogenic, it will
 compete with estradiol for binding to receptor sites, and a classical
 competitive inhibition curve can  be obtained.  From this curve a
 relative  binding affinity (RBA)  can be calculated  that reflects the
 agonistic or antagonistic activity of the toxicant. Such estimates of
 potential estrogenicity may  be used to extrapolate estrogenicity in
 humans and be of importance in approximating the relative risks in
 humans. Although this test  is simple and requires little expense in
 terms of number of  animals, etc., not  all laboratories routinely
 perform such analyses.  However, it is becoming  more  and more
 common and may be standard procedure in the future.
    The major qualifier to such cytosol receptor assays is that certain
 estrogenic compounds, such  as clomiphene and nafoxidine, exhibit a
 very  low RBA-and yet are more estrogenic than predicted (10). In
 part  this is due to the slow clearance of such compounds, which
 provides a  longer  exposure  time and  increases the receptor occu-
 pancy in vivo  when compared to  that  of more rapidly cleared
 estrogens. To detect such long-acting estrogens, estrogen receptors
 assays  can  be  done  in vivo.  Mentioned in  the section  titled
 Qualitative   Reproductive Toxicity  Screen,  these  assays involve
 injecting various dose levels  of the compound in immature rats and
 measuring  the nuclear accumulation and  cytoplasmic depletion of
 estrogen receptors. Reliable,  easy to perform, and sensitive, this test
 requires relatively few animals. It has the disadvantage of not being a
 standard assay in all laboratories.
    Such receptor assays can  be valuable  in  the estimation of
 estrogenic potency  in humans;  however, the chief value of the
 receptor assay probably lies in its ability to  detect estrogen agonist or
 antagonist and has the potential  of elucidating primary steps in the
 mechanism  of  action  of such  compounds.  Such  insights  into
 mechanisms may make future predictions of estrogenic toxicity a
 relatively simple task.

 Androgen agonist—antagonist

    Qualitative screening tests for  androgen  activity  include the
ventral prostate gland hypertrophy produced by  administration of
compounds  to immature (28-day) male rats.

-------
                                20

   Additional models are necessary to evaluate androgenic effects in
the following circumstances:  (a) inhibition of adult female reproduc-
tive function (e.g., ovulation, behavior), and (b) masculinization of
female phenotype (fetal  differentiation,  prepubertal development,
and adulthood phenotypic transformations).
   Inhibition of adult female reproductive function.  The common
clinical response  to  hyperandrogenic  stimuli,  is anovulation. In-
creasing  duration  of exposure or potency  of the agent leads to
oligomenorrhea and secondary amenorrhea. Subtle intensifications of
libido  are experienced by some women,  particularly  with more
potent agents.
    Laboratory testing of adult female rats requires daily evaluation
of vaginal smears for no fewer than four cycles to detect interruption
of the estrous cycle. A daily  1-mg dose  of testosterone propionate
produces diestrus within two  cycles. Appropriate dose-response
studies are indicated.
    Masculinization of the female phenotype: fetal.  After 16 days
of gestation, transplacental transfer  of  potent androgen agonists
results in a variety of imprinting and masculinizing responses that are
based  upon  "critical  periods" of  organ system  differentiation.
Permanent  alterations  in the neuroendocrine regulation  of the
estrous  cycle  and male-type  mating behavior are  "imprinted"  at
lower doses of androgen than are required for disturbing reproduc-
tive tract (vaginal opening) and hepatic monooxygenase (steroid
hydroxylase or dehydrogenase)  activities. A  5-mg dose  of  testos-
 terone propionate administered to the  pregnant dam daily from day
 16 to day 20 of gestation produces the masculinization response in
 female progeny and  does not significantly disturb male differentia-
 tion.  Treatment of neonatal female rats (day  1-10) with a single
 1-mg  dose of testosterone propionate  masculinizes the hypotha-
 lamic-pituitary-ovarian  axis  (persistent estrus)  and sexual behavior
 (male-type with great reliability).
    Masculinization  of  the  female  phenotype:   postnatal  ani-
 mals.  Masculinization  of the female phenotype and suppression of
 female sexual behavior and of the  pubertal events is not induced
 permanently by treatments initiated after the postnatal period (days
 1—10).  Such  masculinization effects  produced in females  tend to
 regress,  and  although vulvar changes may persist, estrous cyclicity
 resumes. Although  these  effects  are clear-cut in rodents,  dose
 extrapolation to humans is not possible.
     Adulthood  phenotypic  transformations.  Masculinization  of
 vulva, mating behavior, and hepatic monooxygenases in adult  animals

-------
                                21

 are much less sensitive indicators of toxicity than the same responses
 in immature animals. Inhibition of ovulation in adult females is also a
 more sensitive indicator of toxicity than the above three parameters.
 Therefore,  additional  tests to assess phenotypic transformation in
 adult females are not necessary.
              Hypothalamic—Pituitary Function Tests

 Assay of agents that stimulate the release of gonadotropins
      from cells of the anterior pituitary gland

    A toxicant may adversely affect reproduction by altering the rate
 of  secretion of one or more  hormones that are synthesized and
 released by the hypothalamus and anterior  pituitary gland. Of the
 hormones  that are  secreted by  the anterior pituitary  gland, the
 gonadotropins (luteinizing  hormone [LH]  and follicle-stimulating
 hormone [FSH]) and prolactin  are most closely  associated with
 reproduction.  The  gonadotropins  are important,  because  these
 protein hormones control  ovarian function, including steroid hor-
 mone secretion,  follicular  development, and' ovulation. Hence,  if
 gonadotropin secretion is suppressed, ovarian function is suppressed.
 A toxicant  could suppress the  secretion of gonadotropins by acting
 directly  on the pituitary gland or by  suppressing the secretion of
 gonadotropin-releasing hormone  (GnRH) by hypothalamic neurons.
    Alternatively,  a  toxicant  could  stimulate  the secretion of
 prolactin, and as a consequence of the  hyperprolactinemia, gonado-
 tropin secretion  becomes  suppressed.  Prolactin  secretion can be
 stimulated by substances that have  estrogenic activity,  substances
 that act as  dopamine  antagonists, substances that inhibit dopamine
 secretion by hypothalamic dopaminergic neurons, or substances that
 cause hyperplasia of prolactin-secreting cells. Some of these actions
 of toxicants can be  assessed (e.g., by quantifying gonadotropin and
 prolactin  secretion),  whereas  others  cannot  be evaluated  in  a
 quantitative sense (e.g., GnRH secretion). A few ways of assessing
 quantitatively  the actions of a toxicant that may have  significant
 effects on reproduction are listed below.
   In vivo  model.  Since agents that stimulate the  release of one
gonadotropin  (e.g., LH) usually affect the release of the other (viz.,
FSH), it is probably only necessary to measure the release of one
(e.g., LH). For such studies,  the estrogen-progesterone-primed female
rat can be used. The Gn-releasing standard should be synthetic GnRH
against which the  test substance can be compared.  The responsive
parameter,  LH in  serum  or  plasma, can be measured  by  a

-------
                                22

standardized radioimmunoassay. After the ED50 of GnRH and the
ED go  of the  test  substance have  been ascertained,  a standard
bioassay can be performed. A three-dose assay, where each dose is
replicated three to four times, may suffice. Thus, such an assay will
require 18 to 24 assay animals.  More animals  can be used if high
precision is  desired. The concentration of LH in plasma or serum can
be evaluated 30 to 60 minutes after the administration of the test
substance and  of the standard. Assuming suitable ranges of concen-
tration and parallel  slopes  from  the two  assays  and a suitable
statistical analysis, such as that described by Bliss (11) for bioassays,
qualitative characteristics of the assay as well as the relative potency
of the unknown  substance  can  be evaluated. If one knows the
potency of GnRH in the estrogenized woman with reference to the
release of LH, it is then possible to calculate the Gn-releasing activity
of the test substance and express the potency in terms of GnRH.
    In  vitro model.  The Gn-releasing  properties of an unknown
substance  can  also   be  measured  using  anterior  pituitary cells
maintained in monolayer culture. In this case the pituitary cells
could be obtained from the rat or a suitable primate. The cells could
be dispersed and established in culture. After three to five days, the
test  substance  and GnRH  can be  assayed  for  their  Gn-releasing
activities, using a bioassay paradigm similar to that outlined above.

Assays of agents that inhibit the release of gonadotropins from
       cells of the anterior pituitary gland

    The details of an in vivo  model assay of a substance that inhibits
 the release  of gonadotropins  could be done as follows. A female rat
 castrated 4 to 6 weeks before testing could serve as the assay animal.
 (In  such an animal,  the  concentration of LH is many times that of
 intact animals.) For a reference standard, 17j3-estradiol could be used
 to suppress the release and hence concentration of LH in serum of
 the test animal. After the ED50 for estradiol and the EDSO for the
 test  substance have been established, a three-dose bioassay could be
 conducted. After an evaluation of the parameters of the  assay, it may
 be possible to calculate the relative potency of the test substance and
 express its potency in terms of 17/3-estradiol.
     After the potency of a test substance relative to 17|3-estradiol has
 been established, one can then calculate the relative potency of the
 test substance in the human by comparing the .LH-lowering effect of
 170-estradiol in castrated or postmenopausal women.

-------
                                 23

Assay of an agent that inhibits the release of prolactin from
      cells of the anterior pituitary gland

    In vivo model.  Although several  experimental animal models
could be used, the young, mature female rat would be adequate. The
only  pretreatment required would be  a short  period (3-4  days)
during which the animal was handled to minimize  the release of
prolactin because of fright. The reference standard for the release of
prolactin  could be haloperidol. Prolactin release could be evaluated
by measuring prolactin in the plasma  or serum of the test animal.
Thirty to sixty minutes after the administration of the test substance
or  haloperidol,  serum or plasma prolactin could be  measured by
radioimmunoassay. After the dose-response curve (i.e., ED50) is
established, a bioassay could be conducted and the relative potency
of the test substance calculated.
    If a dose-response curve for haloperidol in women is established,
one could  approximate the potency of the test substance relative to
haloperidol.  Of course, other reference standards could be used in
this prolactin release  assay  in  vivo.  These include thyrotropin-
releasing factor and vasoactive intestinal peptide.
    In vitro  model.   The ability  of test substance to simulate  the
release of prolactin from  pituitary cells could be conducted in vitro
using pituitary  cells  maintained in monolayer culture.  The donor
could be  the rat as  well as a primate. After a  few (3—5)  days in
culture, a suitable bioassay could be performed,  and the potency of
the test substance relative to a standard could  be evaluated.

Assay of an agent that inhibits the release of prolactin from
      pituitary cells

    In vivo model.  An estrogen-primed female rat could be used in
this assay.  In such an animal the plasma concentration of prolactin is
very high. For  a reference standard, bromoergocriptine could be
used.  After dose-response curves for bromoergocriptine and for the
test substance had been established, a bioassay for prolactin release
inhibition could be performed, where the serum  or plasma prolactin
concentration is  the responsive variable. After a  suitable statistical
analysis,  one could  calculate  the relative  potency  of the  test
substance.  As discussed above, if the dose-response relationship  for
bromoergocriptine in the woman (perhaps an estrogenized woman)
were known, the approximate potency  of the test substance relative
to bromoergocriptine  could be calculated.
    In  vitro  model.  Anterior  pituitary tissue  from  estrogenized
female rats can be used under in vitro conditions to test a substance

-------
                               24

for inhibition of prolactin release. Anterior pituitary tissue can be
incubated  in the presence  of various  concentrations  of bromo-
ergocriptine  or  of the  test substance to establish a dose-response
curve.  Then, using this system, a bioassay can be conducted where
the  concentration  of  prolactin  in the  culture  medium  is  the
responsive variable. Pituitary cells maintained in monolayer culture
could also serve as a suitable in vitro assay system.

Assay of the activity of an agent that alters the secretion of
      dopamine by hypothalamic neurons

    There is no method for the quantification  of  the secretion by
dopaminergic neurons in the human. Although there is a method for
the  measurement of the secretion  of  dopamine into  hypophysial
portal blood, the procedure is  tedious and requires the aid of highly
skilled  people.  Hence, this procedure is not practical  as a routine
procedure. Therefore, one is reduced to making turnover measure-
ments, but such measurements are also  susceptible to large error and
require many animals. Thus, it is reasonable to conclude that as a
routine matter, the rate of secretion of dopamine by neurons of the
brain can not be done for toxicants. This is not to infer that this is
not an important  aspect  of brain  function. Indeed, it is already
known that the secretory activity by dopaminergic  neurons  is
quickly, markedly, and sometimes permanently affected by a variety
of toxicants. Since dopaminergic neurons constitute an important
subset of the neurons of  the  brain, we encourage research on this
important topic.

 Assay of the activity of an agent that alters the secretion  of
       norepinephrine by hypothalamic neurons

     Comments  made  about the secretion of dopaminergic neurons
 are equally applicable to neurons that secrete such biogenic agents as
 norepinephrine  and serotonin. The available techniques for the
 quantitative  study   of neurons secreting  these  agents  are not
 sufficiently advanced to enable their use in routine assays.

 Assay of the activity of an agent that alters the secretion of GnRH

     There is no suitable assay for such an agent at this time.

-------
                                25

 Assay of the activity of an agent that alters the secretion of
      hypothalamic opioid peptides

    It is now clear that morphine and opiatelike peptides affect the
 secretion  of dopamine by hypothalamic neurons and of LH by the
 pituitary gland. Thus, it is  easy to infer an important role for the
 naturally  occurring  opiatelike  peptides  as well  as  morphine in
 reproduction.   However,  this field  is too  new  to  address  in  a
 quantitative manner or to include in a screening system. Yet it can be
 anticipated  that  at   some  time in  the reasonable future,   this
 shortcoming in our technical capabilities will be  surmounted  and
 these problems addressed in  quantitative terms.

 Blood flow of the hypothalamic-hypophysial system

    Perhaps no structure in the mammal has a more complicated
 vasculature  than the hypothalamic-hypophysial complex, consisting
 as it  does of one  component that has a high rate of perfusion  and
 another that is avascular. Moreover, the coexistence in the pituitary
 stalk  of portal vessels carrying blood to the anterior lobe  of the
 pituitary from  the hypothalamus and of a subependymal plexus in
 which pituitary hormones can  pass retrograde in  the stalk  to  the
 hypothalamus attests to the importance of blood flow in this area.
 The  measurement  of blood flow  to the neurohypophysis (i.e.,
 medium  eminence and pars nervosa)  can be measured  accurately
 using radiolabeled microspheres. Blood flow in the anterior lobe of
 the pituitary can be measured using the hydrogen electrode. Thus, an
 area deserving of attention for effects  of environmental toxicants is
 the hypothalamic-hypophysial complex.

 Sexual behavior tests

    Introduction.  Alterations of mating behavior in the female rat
 can  be used   as  an  indicator  of hypothalamic  function and/or
 impairment  of function. A voluminous literature indicates  that
 hypothalamic neurons  serve  as target cells for the ovarian hormones,
 especially  the estrogens,  and that destruction of specific regions of
 the hypothalamus leads  to  abolition  or  disorganization of female
 sexual behavior (12). The studies carried out on the disturbance of
 sexual behavior associated with hypothalamic damage establish three
points of significance  to  the use of the  proposed  sexual behavior
 tests:  hypothalamic  damage can disrupt sexual  behavior  without
altering the  neural systems that mediate pituitary-ovarian function,
the disruption  of  sexual  behavior following hypothalamic damage

-------
                               26

cannot  be reversed  with endocrine  therapy,  and the sexual dis-
ruptions show extensive phylogenetic continuity.
   For these reasons, the sexual behavior system may be capable of
detecting chemically induced abnormalities in hypothalamic function
that  cannot be detected by the other testing systems proposed. Ly
substituting the putative toxicant  for estrogen or progesterone in a
standardized behavioral assay, it is possible to assess its estrogenic or
progestogenic activity in  hypothalamic regions other than those that
modulate pituitary release of gonadotropins. Neural systems modu-
lating female sexual behavior are  by no means limited to hypotha-.
lamic structures. The tests proposed here, however,  are oriented
toward  behavioral end points generally accepted as involving  the
hypothalamus.  A more detailed discussion  of other aspects of sexual
function  and the potential  toxicant-induced  disruption of  other
neural systems is available in Chapter 5.
    Behavioral  assay  methods.  Female  rat  sexual  behavior  has
several  components  that vary, in  a dosage-dependent  manner with
estrogens  and  progestins.  To determine whether   the  toxicant
possesses estrogenic or progestagenic action, the experimenter would
vary the  dosage of the toxicant,  substituting it  for either estradiol
benzoate (EB)  or progesterone in the standard protocol of a behavior
assay. The lordosis response, which includes arching of the back (13),
and the number or latency of approaches  that the female makes to
the  male (14) can be readily quantified.  The testing arena should
have an area of at least four square feet and contain a simple barrier
or compartment.  The females used in the tests should be  ovariec-
tomized  and administered estrogen  and  progesterone (or the sub-
stituted toxicant)  at times to produce mating during the dark phase
of illumination. The estrogen EB (or its substitute) is administered
44 to  46 hours before the  administration of progesterone (or its
substitute), and the  mating behavior is observed  approximately four
hours after administering progesterone.
    A standard dose-response curve  for EB would be obtained by
holding the amount  of  progesterone constant at approximately 0.5
mg and varying the  dosage of EB from 0.1  to 100 jitg. When possible,
the vehicle for delivering these hormones should be the same as that
to be  used  for the  toxicant  and  appropriate  standard  curves
established. The progesterone standard dosage-response curve  would
 be obtained by maintaining EB  at a constant level (5-10 Mg) and
varying the amount  of  progesterone from 0.1 to 10 mg. No fewer
 than 10  subjects can be used to establish the behavioral response
 value at  each  dosage. Repeated mating of the same female at each

-------
                                27

 dosage may be more sensitive than  using different females,at each
 dosage, but each mating should be separated by approximately 5 to 7
 days.
    A reliable method  to determine  the dose-response  curve for
 antiestrogenic activity would be  to use guinea pigs as subjects and to
 measure their lordosis response  to the touch of the experimenter;
 running the index finger along the back, starting from its most caudal
 point.  The  procedure  would  consist of  administering  constant
 dosages of EB (5-10 pig) and, 46 hours after the initial dose of EB, a
 0.5-mg dose of progesterone. When administered simultaneously with
 estrogen, progesterone  possesses antiestrogenic  properties in  the
 guinea pig  and can  therefore be used as a standard to assess the
 antiestrogenic activity of the toxicant.  Accordingly, varying dosages
 of the compound (0.1—10 mg) would be administered along with the
 EB.
    The antiestrogenicity  of a putative toxicant can also be evaluated
 in the female rat's behavior system. However, because this behavior
 system is  relatively  insensitive  to the inhibitory actions or anti-
 estrogenic  actions of progesterone, an alternative antiestrogen, such
 as MER-25, is recommended for a comparative standard.
    The toxicant can also be administered in addition to the standard
 doses of EB or progesterone.  This can be done at the same time that
 estrogen and progesterone are administered to determine whether it
 potentiates  or antagonizes the action of each of these steroids. In
 addition, the toxicant can be administered prior to the determination
 of a  standard dose-response curve, and the  estrogen dose-response
 curves can be compared with those obtained from control animals. In
 this way, toxicant-produced modifications of the brain areas medi-
 ating sexual behavior can be detected using a test based on a standard
 estrogen dose-response curve.
   Relevance to humans. A positive result  from these behavioral
screening procedures could reflect disruption of neural, most likely
hypothalamic, function and could indicate the potential for inter-
ference with human hypothalamic function. However, the manifesta-
tions of this potential hypothalamic disruption in humans will most
likely be different from those in rodents.
                        Ovarian Toxicity

Oocyte and follicle toxicity

   The ovary is responsible for two roles in reproduction: nurture
and  release of  gametes  and  hormone production.  Clinical and

-------
                                28

experimental data demonstrate that a variety  of xenobiotic com-
pounds can alter both aspects of ovarian function (15, 16). Multiple
studies  have demonstrated that one of cigarette smoking's adverse
effects  on  the human ovary  is  an earlier  dose-related age  of
menopause. The assays described  here are  designed  to assess the
effect of xenobiotics on the  first aspect of ovarian function, gamete
nurture. Tests for xenobiotic effects on oogenesis can be determined
by  including prenatal as well as postnatal  treatments. Xenobiotic
destruction of oocytes is of great  significance because the effect is
irreversible: there is no mechanism for repopulation of oocytes in the
ovary.
    Evidence suggests that inbred mouse strains represent the most
sensitive test strains for oocyte and follicle toxicity assays  (17).
Additional data in other species and with other xenobiotics is needed
to  clarify  this relationship. Inbred mouse strains also  offer the
advantage that they provide  the logical framework for exploration of
the mechanism of action as well as providing a reproducible assay
system (18-20).
    After  treatment  with   the  compound  of interest,  mice  are
sacrificed at varying time intervals  and their ovaries removed, fixed,
serially sectioned, and stained. Oocytes and  follicles are quantitated
using  a microscope, and effect  of treatment on  oocyte or follicle
number is  determined. Follicles and oocytes are classified by  the
method of Zuckerman (21). This assay, although cumbersome, is
easily learned and conducted by laboratory technicians.
    Evidence  suggests  that  this assay  is a much more sensitive
indicator of oocyte or follicle damage, than alterations in fertility.
Unpublished investigations at the Pregnancy Research Branch of the
National Institute of Child Health  and Human Development, as well
as other published data, suggest that as many as 90% of all oocytes
have to be destroyed before short-term alterations in  fertility of the
female can be observed.
    The full  range  of specificity  of  the assay  has yet  to be
determined. The assay  appears  to  be  quite sensitive  and dose
dependent (see Table 4 for available ED5 0 's).

 Inhibition of steroidogenesis
     In developing a model system to estimate the quantitative risk of
 a  toxicant  with regard to inhibition  of  ovarian steroidogenesis,
 multiple physiological  and technical aspects must be  considered.
 These  include (a) the cell-type specific sex steroids to be measured,

-------
                                          29
Q  £P
W  g

     CD
Compound
                  A  A  A  A
                            — i

                            g
                                                                         ^
                                                                     Cq  ^
                                                                    ti  o
                                                                     CQ  +•>
                                                                     c  c
                                                                     0)  o
                                                                     8  5
                                                  o   o   o   o  .ti  !3  -tl
                                                  •*-<  •»-!  -T-l  «1H   S"*  ?  _Q
              OOOOOOOOOOOOo45
                                                                                       CO
                                                                                       s
                                              c
                                              03


                                              CD
                                              CO
                                s
                       CD  r-<
                       c  §
                       (U  )H
CD

CD


                      -5 S9 -5   «   -S
                                             '

                                                               1

                                                                                      s
                                              CO

                                             1
                                             £

                                             I
                                              V3
                                              CD

-------
                                30

(b) the  cooperative compartmental  steroid  biosynthesis  charac-
terizing the follicular phase, (c) the cycle-related variations in sex
steroid  production, (d) the key regulatory steps in steroid bio-
synthesis (i.e., the availability of substrate and the roles of luteinizing
hormone  [LH]  and follicle-stimulating hormone [FSH]),  (e) the
available methodology, and (f) the ability to extrapolate the data
between species. The focus here is placed on a model in vitro rather
than in vivo because of considerations regarding the specificity of the
toxic effect, the lack of interference by nervous or humoral factors
present in vivo, the greater likelihood of intracellular interaction with
the toxicant,  the applicability of the test system, and the ability  to
extrapolate the data to human or at least primate ovarian cell types.
    Estrogen,  primarily  17/3-estradiol progesterone, 17a-OH proges-
terone, androstenedione,  and  testosterone, are  the  predominant
steroids produced by the human ovary during the reproductive years.
Estrogen characterizes the follicular phase,  with  the corpus luteum
producing  both  estrogen  and  progesterone and a drop  in both
steroids occurring  at the time of menses in a nonconceptive cycle.
Androgens  are secreted throughout a nonconceptive cycle, with a
slight rise at midcycle. Controversy  still exists concerning the cell(s)
of origin of follicular estrogen; both direct thecal cell secretion and
granulosa cell aromatization of thecal androgen are supported in the
literature (23). Because granulosa  cells lack  the 17,20 desmolase
enzyme, the  thecal and interstitial  compartments are felt to be the
source of Ct 9 androgens.  After ovulation the granulosa and thecal
compartments both form  the  corpus luteum and produce proges-
terone and estrogen. Any  model system using ovarian cell  types in
vitro must consider these differences as well as the  overall cyclic
steroid secretory pattern characteristic to the species utilized.
    Regulatory steps in gonadal  steroid secretion include  (a) sub-
strate (cholesterol)  availability  (i.e., the  low-density  lipoprotein
fraction of plasma); (b) luteinizing-hormone (LH) induction of the
20,22-hydroxylase-desmolase steps converting cholesterol  to preg-
nenolone,  and (c)  follicle-stimulating-hormone (FSH) induction of
granulosa  cell aromatase  activity  converting thecal  androgens to
estrogens.  Since thecal steroidogenesis has not been demonstrated to
depend on FSH-induced aromatization, LH stimulates thecal  andro-
gen production,  and low levels of LH are required in  vivo for
adequate luteal  function,  some  of these regulatory steps  may be
compartment specific.
    A toxicant may not demonstrate inhibition of steroidogenesis in
vitro and yet be active in vivo, if it affects selectively gonadotropin-
mediated events in vivo or only progesterone synthesis stimulated by

-------
                                 31

 human .chorionic  gonadotropin  (HCG)  (24, 25), and hence  be
 detectable only in vivo in a known conceptive cycle. Similarly, agents
 acting through prostaglandins known to induce luteal regression in
 vivo in some species (26) may be active only in  vivo, because the
 agents may act not directly  on the steroid-secreting cell but rather
 indirectly by selective ovarian veno-constrictive action. Despite these
 possibilities,  most  known   inhibitors  of  steroidogenesis  act  by
 affecting specific enzymes in the steroid pathways (Table 5).
TABLE 5 Agents That Inhibit Steioidogenesis
Steroidogenic Step
20a hydroxylase
Sidechain cleavage
Dehydrogenase, 3(3-hydroxy-
A* -steroid


Aromatase


11/3-hydroxylase


21-hydroxylase
17a-hydroxylase


17,201yase
' Inhibitor
Amino-glutethimide phosphate
3-methoxybenzidine
Cyanoketone
Estrogens
Azastene
Danazol
4-acetoxy-androstene-3,17-dione
4-hydroxy-androstene-3,17-dione
l,4,6-androstatriene-3,17-dione
Danazol
Metyrapone
SKF-12185
Danazol
Danazol
SU-9055
.SU-8000
Danazol
    Adequate methodology is currently available for (a) isolation of
ovarian cell types (27, 28), (b) tissue or organ culture, and (c) direct
radioimmunoassay of media for individual steroids without chroma-
tography steps. If HCG stimulation of steroidogenesis is required to
demonstrate an effect, serum-free media may be required, as there is
some evidence that blocking factors for gonadotropins are present in
serum (29); but in short-term cultures (<24 hours), the lack of serum
factors  should not present a  problem  for cell viability.  Plating
efficiency can be determined by supravital staining, and  cell counts
or determinations of DNA or protein can be used to normalize data.
Organ cultures are  more difficult to  normalize because of  more
heterogeneous cell populations, less well-defined culture conditions,
and more difficult assessments of cell viability, but tissue wet weights
can be used. Enzymatic dispersion techniques are available (25), but

-------
                                32

they add considerable time to the procedure and do not solve the
problem of cell heterogenicity. Furthermore, if gonadotropin stimu-
lation  is  required, highly purified  enzyme preparations  (i.e., col-
lagenase)  are necessary to avoid protese contamination and altera-
tions in membrane-bound protein receptors (30). Hence, because of
ease of culture, purity of cell type, and active basal steroidogenesis,
isolated granulosa cell cultures, with or without added Ci 9 androgen
substrate, represent attractive models for evaluation of a potential
toxicant's effect on steroidogenesis (Table 6).
    Cell-cell interactions may control the pattern of ovarian steroido-
genesis as evidenced by  the  so-called "spontaneous luteinization"
that granulosa cells undergo when placed in  tissue culture inde-
pendent of when they are harvested in the follicular phase (31). The
removal of the cells from their  approximation to the thecal layer,
contact  with  follicular fluid, or disruption of intimate cell-to-cell
contact appears to alter their steroidogenic potential and morpho-
logic appearance  in vitro. For these reasons the use of intact follicle
walls without separation of the  thecal and granulosa compartments
may  have to  be  considered as  a  test system if problems  are
encountered with isolated cell systems.
    Selection of the species for use depends primarily on availability
of adequate numbers of physiologically matured follicles or corpora
lutea.  While diethylstilbestrol-treated immature rats can be used as a
source of ovarian cells (32), the numbers of cells  are  small and
require  a substantial  time  investment  for  collection.  Domestic
animals,  by  contrast,  have much larger follicles, and the use  of
slaughterhouse material of a polyovulatory  species minimizes  the
precollection time investment. Pigs and cows are  the most desirable
large  animals  to use in  this regard, and both  have an extensive
literature available regarding their reproductive cycles, cell collection
techniques, and tissue culture. The cell system chosen should be an
easily  exploitable model  system  in which  known  inhibitors  of
steroidogenesis in both human and animal systems can be studied in
vitro  to  validate the animal model  and the  data extrapolated  to
humans for more general application  to quantitative risk assessment
of other suspicious compounds.

-------
33













.S
S
D


1
o
g"S>
•- S
*• M
tr'g
c B
o •*j
•| co
H O
o, c
S .2
B* .IS
§1
atures of Ovarian
Xenobiotic )
i
H








s a
r—t •*-> ^
E B S
2 S u
•M ^
00 M





•a  a
<§ S




/*S
« o <3 j
OJ oo •*-* i i , •<: i
gas ' ' + z '
B| g
w < £








•**» §
O S) "« 1 + + 1 1
III
<: w £
C rS "M1
o 4,, -g S
S! S> •& !§ 'S 1 1 + + +
11111
BH W

bO .£H

"° n C
•§ S c & c S 3
2-a .8 "H .8 ™ o
gis's a'iaSo'i "i
•Soo C^Sci^'O^S .52
£"ai£ T3Ca-*ag-M <*-<
ia^(+_, agSo^'a*^^
<2 00 00 < (2 (S





o
3
s
a
0.
S"
o
c
i
I
Z
c
1
1 d .
.1 -1
tin O
.s S
•0 >.
1 §
o *u
**3 «H

*G cf
"o *^
C3 g>
c3 3
•S i
« "
"SB §
nus(-), absence
varying proportio
roportion.
itic digestion; org
•= _ cu £
S.S M E
« 2 .S N
1-2 l§
3 "3 g K
8 S S-§
B <> 3

2 2 o M
01 "c S a
S « "> 3
s «11
III!
•i- S ° S
S|1 |
•3-S fi.f
< H W Q
a -Q y ^3


-------
                                   34
                            REFERENCES

 1. Gellert, R. J., Bakke, J. L. and Lawrence, N. L.: Persistent estrus and altered
    estogen  sensitivity in  rats treated neonatally with  clomiphene  citrate.
    Fertil. Steril. 22: 224-250, 1971.
 2. Clark, J.  H. and  McCormack, S. A.: The  effect of  clomid  and other
    triphenylethylene derivatives during pregnancy and the neonatal period. J.
    Steroid Biochem. 12: 47, 1980.
 S.Eroschenko, V. and Palmiter, R.: Estrogenicity  of kepone  in birds  and
    mammals. In: Estrogens in the  Environment, J. McLachlan,  Ed., Elsevier/
    North Holland: New York; pp. 305-326, 1980.
 4. Clark, J. H. and  Peck, E. J., Jr.: Female  Sex  Steroids:  Receptors  and
    Function: Springer-Verlag: Berlin; 245 pp., 1979.
 S.Katzenellenbogen, J., Katzenellenbogen, B., Tatee, T., Robertson, D. and
    Landratter, S.: The chemistry of estrogens and antiestrogens. In: Estrogens
    in  the  Environment, J. McLachlan, Ed., Elsevier/North Holland: New
    York; pp. 33, 1980.
 6. Kupfer, D. and Bulger, W.: Estrogenic properties of DDT and its analogs. In:
    Estrogens in the Environment, J. McLachlan, Ed., Elsevier/North Holland:
    New York; pp. 239-263,1980.
 7. Chan, L.  and O'Malley, B. W.: Mechanism of action of sex steroid-hormones.
    I. N. Engl. J. Med. 294: 1322-1328, 1976.
 8. Chan, L.  and O'Malley, B. W.: Mechanism of action of sex steroid-hormones.
    II. N. Engl. J. Med. 294: 1372-1381, 1976.
 9. Chan, L.  and O'Malley, B. W.: Mechanism of action of sex steroid-hormones.
     HI. N. Engl. J. Med. 294: 1430-1437,1976.
10. Clark,  J.  H., Anderson,  J.  N.  and Peck, E. J.,  Jr.: Estrogen receptor
     antiestrogen complex: atypical binding by uterine nuclei and effects on
     uterine growth. Steroids 22: 707-713, 1973.
11. Bliss, C.  I.: Statistical  methods in vitamin research. In: Vitamin Methods,
    P. Gyorgy, Ed., Academic Press: New York; pp. 448-609, 1951.
12. Pfaff,  D.  W.:  Estrogens  and  Brain Function:  Neural  Analysis of  a
     Hormone-Controlled Mammalian Reproductive Behavior. Springer-Verlag:
     New York; 272 pp., 1980.
IS.Gerall, A. A.  and McCrady, R. E.: Receptivity scores  of female   rats
     stimulated either manually or by males. J.Endrocrinol. 46:  55—59, 1970.
14. McClintock, M. K. and Adler,  N. T.: The  role of the female  during
     copulation in  the wild and domestic Norway rat.  Behavior 67(1—2):
     67-96, 1978.
15. Mattison, D. R.: How xenobiotic compounds can destroy oocytes. Contemp.
     Ob.Gyn. 15: 157-169,1980.
16. Mattison, D. R. and Thorgeirsspn, S. S.: Smoking and industrial pollution
     and their effects on menopause and ovarian cancer.  Lancet 1: 187—188,
     1978.
17. Mattison, D. R.:  Difference  in  sensitivity  of rat and mouse primoridal
     oocytes to destruction by polycyclic aromatic hydrocarbons. Chem. Biol.
     Interact. 28: 133-137, 1979.

-------
                                    35

 18. Mattison, D. R. and Thorgeirsson, S. S.: Gonadal arylhydrocarbon hydroxy-
      lase in rats and mice. Cancer Res. 38: 1368—1373, 1978.
 19. Mattison, D. R. and Thorgeirsson, S. S.: Ovarian arylhydrocarbon hydroxy-
    lase activity and primordial occyte toxicity of polycyclic aromatic hydro-
    carbons in mice. Cancer Res. 39: 3471-3475, 1979.
 20. Mattison,  D.  R.,  West, D.  M.  and  Menard,  R. A.:  Differences  in
      benzo(a)pyrene  metabolic profile  in  rat  and  mouse ovary. Biochem.
      Pharmacol. 25: 2101-2104, 1979.
21. Zuckerman, S.: The Ovary. Academic Press: New York; 600 pp., 1962.
22.Pedersen, T. and Peters, H.: Proposal for a classification of oocytes and
      follicles in the mouse ovary. J. Reprod. Fertil. 17: 555-557, 1968.
23. Armstrong, D. T. and Dorrington, J. H.: Estrogen biosynthesis in the ovaries
      and testes.  In:  Regulatory Mechanisms Affecting  Gonadal Hormone
      Action, Advances in Sex  Hormone Research, J.A.  Thomas,  and R. L.
      Singhal, Eds., University Park Press: Baltimore; 3: 217-258, 1977.
24. Stouffer, R. L., Nixon, W. E. and Hodgen, G. E.: Estrogen inhibition of basal
      and gonadotropin-stimulated progesterone production by Rhesus monkey
      luteal cells in vitro. Endocrinol. 101: 1157-1163,1977.
25. Williams, M. T., Roth, M. S., Marsh, J.M. and LeMaire, W. J.: Inhibition of
      Human chorionic gonadotropin-induced progesterone synthesis by estra-
      diol in isolated human luteal cells.  J. Clin.  Endocrinol.  Metab. 48:
      437-440, 1979.
26. Goldberg, V. J. and Ramwell, P. W.: Role of prostaglandins in reproduction.
      Physiol. Rev. 55: 325-351, 1975.
27. McNatty, K. P., Makris, A., DeGrazia, C., Osathanondh, R. and Ryan, K. J.:
      The production  of  progesterone, androgens and estrogens by granulosa-
      cells, thecal  tissue, and  stromal tissue from human ovaries, in vitro. J. Clin.
      Endocrinol. Metab. 49:  687-699, 1979.
28. Haney, A. F. and Schomberg, D. W.: Steroidal modulation of progesterone
      secretion  by granulosa-cells  from  large  porcine  follicles:  a role for
      androgens and estrogens in controlling steroidogenesis. Biol. Reprod. 19:
      242,1978.
29. Erickson, G. F., Wang, C. and Hsueh, A. J. W.: FSH induction of functional
      LH receptors in granulosa cells cultured in a chemically defined medium.
      Nature 279: .336-338,  1979.
30. Gulyas, B. J.,  Yuan, L. C. and Hodgen, G. D.: Progesterone production by
      dispersed monkey (Macaca Mulatto) luteal cells after exposure to trypsin.
      Steroids 35:  43-51, 1980.
31. Channing, C. P.: Influences of the in vivo and in vitro hormonal environment
      upon luteinization of granulosa cells in tissue culture. Recent Prog. Horm.
      Res. 26: 589-622, 1970.
32. Hall, P. F. and Young, D. G.: Site of action of trophic hormones upon the
      biosynthetic pathways to steroid hormones. Endocrinol. 82: 559, 1968.

-------

-------
             II. GLOSSARY OF TERMS USED IN
                 FEMALE REPRODUCTION

 adenosis—a nonneoplastic  glandular disease  that  occurs in the
 uterine arnix and upper vagina.

 amenorrhea—absence or abnormal cessation of the menses.

 androgen—a class of steroid hormones produced in the gonads and
 adrenal  cortex that regulate masculine sexual characteristics; a
 generic term  for agents that encourage the development of or prevent
 changes in male sex characteristics; a precursor of estrogens.

 androgen antagonist or antiandrogen—agent that opposes or im-
 pedes the action of an androgen.

 anovulation—suspension or cessation of the escape of ova from the
 follicles.

 corpus  luteum—an endocrine body  formed in ovary  at site of
 ruptured  Graafian  follicle  that. secretes an estrogenic and pro-
 gestagenic hormone.

 diestrus—quiescent  period following ovulation in the estrous cycle
 of female mammals  in which the uterus prepares for reception of a
 fertilized ovum.

 dopamine or hydroxytyramine—an intermediate in tyrosine catabo-
 lism and the precursor of norepinephrine and epinephrine.

 ectopic pregnancy—pregnancy occurring outside the uterine cavity
 egg—female sexual cell.

 estradiol—an estrogenic hormone (C18H24O2) produced by follicle
 cells of the vertebrate ovary; provokes estrus and proliferation of the
human endometrium,

estrogen—estrogenic hormone; generic term for various natural or
synthetic substances that produce estrus.

estrogen agonist—an agent that has a biological activity similar to
that of the physiological estrogens.
                              37

-------
                               38

estrogen antagonist or antiestrogen—agent that opposes or impedes
the action of an estrogen.

estrus—phase of the sexual cycle of female mammals characterized
by willingness to mate and in intact animals when ovulation occurs.

follicle  (ovarian)—one of the vascular  bodies in the ovary, con-
taining the oocytes.

follicle-stimulating  hormone  or FSH—a  glycoprotein  hormone
secreted by the anterior  pituitary  of  vertebrates  that  promotes
spermatogenesis and stimulates growth and secretion of the Graafian
follicle.

galactorrhea—continued discharge of milk from  the breasts in the
intervals between nurshing or after weaning.

gonadotropin—a substance that acts to stimulate the gonads.

Graafian follicle—mature mammalian ovum  with its surrounding
epithelial cells.
                                                   s
gynecomastia—excessive development of the male mammary glands,
sometimes leading to milk secretion.

hypothalamic-pituitary-ovarian  axis—the hormonal interactions that
link and control female reproduction.

hypothalamic-hypophyseal complex—the  structural and  hormonal
relationships between the hypothalamus and the pituitary.

lactation—the production of milk;  the  period following  childbirth
during which milk is formed in the breasts.

luteinizing hormone or LH—glycoprotein hormone secreted by the
adenohypophysis of vertebrates that stimulates hormone production
by interstitial cells of gonads.

menopause—natural  physiologic  cessation of menustration,  nor-
mally occurring in the last half of the fourth decade.

oligomenorrhea—prolongation of menstrual  cycle  beyond average
limits.

-------
                                39

 oocyte—female ovarian germ cell present after birth.

 ovarian  cyclicity—the  periodic changes  observed in  the  ovary
 associated with follicular growth,  ovulation,  and corpus  luteum
 function.
 ovulation—discharge of an ovum or ovule from a Graafian folicle in
 the ovary.

 parturition—labor; giving birth.

-postpartum estrus—estrus  with ovulation and corpus luteum pro-
 duction  which occurs in some species immediately after  birth of
 offspring.

 progesterone—a steroid hormone (C21H3oO2) produced in corpus
 luteum, placenta, testes, and adrenals that plays a physiological role
 in the luteal phase of menstrual cycle and maintenance of pregnancy;
 also an intermediate in biosynthesis of androgens and estrogens.

 prolactin—a protein hormone produced by adenohypophysis that
 stimulates secretion of milk and promotes functional activity  of the
 corpus luteum.

 prostaglandins—various 20-carbon-atom compounds, formed from
 essential fatty acids, that physiologically affect the female reproduc-
 tive organs, the nervous system, and metabolism.

 puberty—period at which the generative organs become capable of
 reproduction.

 relative binding affinity—the degree to which a ligand, compared to
 standard ligand, is bound to a receptor.

 secondary amenorrhea—any case in which the menses appeared at
 puberty but have been suppressed.

 steroidogenesis—enzymatic steps converting acetate and cholesterol
 to sex steroids, glucosteroids, or mineralocorticoids.

 testosterone—a biologically potent androgenic steroid which may be
released from the gonads and adrenal glands.

-------
                               40

virilization or masculinization—the assumption of male characteris-
tics  by a  female because  of excessive production of androgenic
substances or masculinizing tumors of the ovaries.

-------
                         CHAPTER 3
   CONSIDERATIONS IN EVALUATING RISK TO

                 MALE REPRODUCTION

                       INTRODUCTION

   During  evolution  the  reproductive patterns  of mammals, in-
cluding man, were determined to a considerable extent by the nature
of the environment. Similarly today a variety of natural environ-
mental factors may alter reproductive activity and fertility. Potential
hazards to man's reproductive state are present in the environment as
pollutants whose effects are often not likely to be clear-cut. Thus,
sensitive assessment systems are needed. However, it is not clear how
the potential effects on male reproduction can best be assessed.
   Detailed information about many aspects of male reproduction
exists, but there is  little firsthand  experience with detection in
animals of  subtle effects of either new chemicals or environmental
hazards. The task of detecting effects that may  be reflected  only
marginally  in fertility performance  is made more difficult  by the
variability of different reproductive parameters such as the concen-
tration of  sperm in an  ejaculate,  the  total number of sperm
ejaculated,  and  the  sperm  morphology  within a  population  of
"normal"  men hi our society.  Hence, our understanding  of the
reasons for this variation and our ability to evaluate subtle responses
to environmental hazards is minimal, and it is important to recognize
the embryonic state of our  abilities in this regard.
   It is clear that changes in human reproductive function induced
by environmental hazards might  be reflected  in reproductive be-
havior; in circulating levels of hormones such as follicle-stimulating
hormone (FSH), luteinizing hormone (LH), and testosterone; or in
testicular and  epididymal  function as evidenced  by spermatogenic
activity, fertilizing potential of the ejaculate, and ability of the sperm
genome to  support normal development after fertilization. Monitor-
ing of a human population  for the normality  of  any  of these
functions and  assessment of the risk of certain levels of a chemical
hazard require objective criteria that are measurable hi man  and,
when a new chemical is to be evaluated,  in an appropriate  animal
model.  Ejaculates of human  and  of  animal  semen contain a
                              41

-------
                               42

considerable heterogeneity of spermatozoa, and several parameters of
ejaculates may vary considerably from sample to sample. Nonethe-
less, there is  now a reasonable knowledge of many facets of the
normal physiology and of the variations to be expected for many
specific parameters in animal models and to a lesser extent for man.
The guidelines described  below are based on current knowledge of
the  physiology  of male reproduction in mammals, including man,
and suggest an approach to risk assessment of existing and potential
chemical hazards for reproductive function.

                     Aspects of the Problem

    The single most  sensitive and  important parameter for human
fertility is the total number of motile sperm in an ejaculate (1, 2). It
has not been possible to set an exact limit on the minimal number of
motile sperm  per  ejaculate, or what is more commonly reported as
concentration of sperm or semen,  necessary for  fertility in man. A
male ejaculating as few as 1 X 106 sperm per milliliter may prove
fertile occasionally (3), but in most cases low numbers of sperm per
ejaculate bear an  obvious relationship to infertility. For example,
sperm concentrations below  10 million, of from 10  to 20 million,
and from 20 to 40 million per milliliter are associated with a risk of
infertility that is, respectively, tenfold, fivefold, and threefold higher
than for individuals with normal spermatozoal concentration,  that is,
60 to  160 million per milliliter (4). Because another study (5) shows
that relative risks  are fourfold and twofold higher for men with
sperm  concentrations of below  10  million and between 10 and 20
million per milliliter, respectively, it is quite possible that a twofold
reduction  in sperm concentration in individuals with sperm counts
below 40 million per milliliter will double the incidence of infertility.
Several characteristics of human semen and testicular function reflect
a low  efficiency (2, 6).  Human testes  may  function often  at the
threshold of pathology (2, 7,  8) and may be particularly sensitive to
toxic agents compared with the testes of animals  commonly used to
study testicular function.
    The yield of spermatozoa from  spermatogonia, the rate of sperm
production per gram of testis, and the percentage of morphologically
normal sperm  in  ejaculates  are lowest in man  among the many
mammals   studied (2,6—8).  The  median  number   of  sperm
(~200 X 106 per ejaculate) is only fourfold higher than the value
(50 X 106 per ejaculate) below which fertility becomes significantly
reduced (9). In contrast, the number of sperm in an ejaculate of bull

-------
                                43

 semen  (7 X 109),  is  1400-fold  higher  than the value of 5 X 106
 sufficient to achieve maximal fertility by artificial insemination (10),
 and a smaller animal,  the rabbit, also shows a large differential. It is
 possible that a given set of conditions in the environment may cause
 infertility in man more readily than in experimental animals. Several
 agents,   including  radiation  (11—14),  chemotherapeutic  drugs
 (15—18), and dibromochloropropane (19—22), reduce motile sperm
 concentrations and affect fertility.


                   Selection of an Animal Model

    Evaluation of  compounds for  potential risk to human males
 requires one or more animal models. The selection and use of these
 models   for testing end points that signify a reproductive hazard
 generally is more specialized than that for most toxicology  or
 mutagenesis testing. The relevant end points depend on integrated
 functional aspects  that can be monitored with ease only in certain
 species.  The use  of two species reduces the possibility of missing a
 hazardous agent during testing.
    Parallel  testing of both  rat and rabbit seems  most  suitable.
 Although a number of laboratory or domesticated species might be
 used, rabbits and rats  offer several advantages in comparison to dogs
 and subhuman primates. Rabbits have a high, predictable libido that
 may be  useful in assessing risks to sexual  behavior.  More importantly,
 all  sequential  phases  of  the conception  process  (i.e., endocrine
 function, spermatogenesis, sperm  maturation, ejaculation,  sperm
 capacitation in the female,  and fertilization) are easily evaluated,
 quantified,  and manipulated throughout the year.  The ability to
 characterize the whole ejaculate quantitatively and qualitatively and
 the ability to collect the ejaculate with ease using an artificial vagina
make the rabbit a key test model for sensitive assessment of possible
harmful effects of environmental agents on male reproduction.
    The rat  is also a very useful model and is preferable to the mouse
or  hamster  because of  the  rat's widespread use in lexicological
research, the large  base  of knowledge of its reproductive processes,
its relatively low cost, its  convenient size for weighing organs,  and
the fact that it breeds  readily under laboratory conditions. The rat is
less  useful  than  the  rabbit,  because more  of  the measurements
require  invasive procedures and/or sacrifice of the individual/ The
characteristics of several  potential models are summarized in Table 7
(1, 23).

-------
44




li
S
1
M
"w
•g
1
•a
a
1
u.
.5
c
o
o
3
O


1
O
_o
a
3
w
M
*2
.g
4>
S
TABLE 7



•1


>» *w5*
"c i
O £
IS £.


2T
o|

T3
« J ^
3 o> 2
<5 a- ^
&
z.




rt
«
s
o
s









0
vd


"0




NO
CO


p.
o






ON
c*i

vo
CO*


tn
3
O
0>
a
.a
of cycle of sei
c
.2
COCSVOONVO CSOCO •<*•
v^^iOC-^ <=>^0^ ^JQS
rH TJ-
o\ o »o
o^o»ooot— T-HOCO
r^voc^«-ic^ O»-O>O ?— iO^J"O ^
•^h in en vo co O* t-J o 04 o O e--
^ -1 ^c^


CO O CO
COCOC"»-HCS OO^TJ"
^c--o'oi +-> rt
«j 3 -O
*8&

-------
45












"CJ
§
.s
s
o
CJ

«
3
H







S
03
5? ^
C cS
o .e
-s-
"> "a
O 5f
Q a>
•a
B
|| 3
a>
z
^"^



+jj
*

a>
o






McAcaco ^^eocnwsoswi
>.>->,>, 2Z>.>,>,>->.
C) ^J
cACAcAco wsertwsvswa
si Sj sf ^ ^izsisJ1^^!^


c« -co co co waOTenwawa

^ ^ ^ ^ ^^^^^


ssss sssssss
>,>.>.>. >H >H





SSSo SjOeSooSo
[>•( ^X )>« £5 J>4 ^ J>-C ^ IZ] ^M J>l


z||z ^z^zz^z


1.1° i

w> P S 3 O «
S S <2 o -c ^
I -a S c .fe " S
1 S « -2 SS>-«
o 1 || | ^-~ig
|lll||j|i.|lll















c
3
_O oi
3. a
S "3
it 3
Q |
1 'i
x'E e .
O o O ^
rt -^3 fS
ro ^ ^
II ,2 3 G
a, o o a
fi «J i

-------
                               46

            Tests for Evaluating Reproductive Damage

   We reviewed a wide spectrum of test systems. Table 8 (24—56)
lists  tests that were  considered to be  suitable for qualitative and
quantitative risk assessment  (for detailed consideration, see the
Appendix to  this chapter). Table 9 (including Refs.  57-70) lists
additional tests that were considered but rejected because they are
insensitive, redundant, not cost effective,  too difficult to perform
unless within a research setting, not validated, too controversial, or in
need of further development.
   The  tests  in Table 8 evaluate the endocrine control of male
reproduction  and the number and quality of sperm  produced,  or
they measure fertility. Since fertility is related to the number  of
normal spermatozoa in the ejaculate, the analyses of seminal quality
are indirect measures of fertility.  However,  comprehensive seminal
analysis is a much more sensitive end point for detection of a toxic
effect than a  breeding experiment using natural mating, because in
experimental animals the number of sperm ejaculated greatly exceeds
the  number necessary for fertility (10,71). The various tests are
stratified into a sequence (tests 1-3, S, E) ranging from a prelimi-
nary screen to more detailed studies.
    Coefficients of variation (CV) might be used to  determine the
sensitivity  of a study that compares treated animals  with controls
(see Appendix). Generally,  measurements  of testicular and epididy-
mal sperm numbers give highly reproducible values in control animals
with coefficients of variation between animals of 15% or less (24).
This degree of reproducibility  ensures that  the  test will be quite
sensitive, even with relatively few animals (Table 10). Although there
is appreciable variation (CV = 70%) in sperm concentration or total
number of sperm per ejaculate in semen  collected, from rabbits or
bulls (72, 73), this variation  can be  reduced somewhat by using a
uniform  interval  between  seminal  collections  and  standardized
procedures in chronic studies. Sperm motility and morphology are
much more constant than sperm number or concentration, especially
within individuals of a species (10, 74). These two assays usually are
performed  in a  subjective  manner, and  efforts must be made to
minimize  this subjectivity  (2).  In some species motility  can  be
 evaluated  objectively by measurements  made  on time exposure
 negatives (75)  or probably  better on videotape recordings (49).
 Morphology  of spermatozoa from  individuals always  should  be
 compared to control samples analyzed concurrently  by the same
 observer.  The  slides and/or videotapes  should  be  retained  for
 validation by  an outside observer.

-------
47
TABLE 8 Tests Considered Useful for Screening Toxic Compounds"
Test
Body Weight
Testis
Size in situ
Weight
Spermatid reserves
Gross histology
Nonfunctional tubules (%)
Tubules with lumen sperm (%)
Tubule diameter
Counts of leptotene spermatocytes
Epididymis
Weight of distal half
Number of sperm in distal half
Motility of sperm, distal end (%)
Gross sperm morphology, distal end (%)
Detailed sperm morphology, distal end (%)
Gross histology
Accessory Sex Glands
Weight of vesicular glands
Weight of total accessory sex glands
Semen
Total volume
Gel-free volume
Sperm concentration
Total sperm/ejaculate
Total sperm/day of abstinence
Sperm motility, visual (%)
Sperm motility, videotape (% and velocity)
Gross sperm morphology
Detailed sperm morphology
Concentration of agent in sperm
Concentration of agent in seminal plasma
Concentration of agent in blood
Biochemical analyses of sperm/seminal plasma
Endocrine
Luteinizing hormone
Follicle-stimulating hormone
Testosterone
Gonadotropin-releasing hormone
Fertility
Ratio exposed: pregnant females
Number embryos or young per pregnant female
Ratio viable embryos: corpora lutea
Ratio implantation: corpora lutea
Number 2-8 cell eggs
Number unfertilized eggs
Number abnormal eggs
Sperm per ovum
Number of corpora lutea
In Vitro
Incubation of sperm in agent
Hamster egg penetration test
Rat
1-3

1-3
1-3
1-3*
1-3C
2,3*
2,3
2,3
1-3

1-3
1-3
1-3
1
2,3
NA

1-3
NA

NP
NP
NP
NP
NP
NP
NP
NP
NP
NP
NP
NP
NP

2,3
2,3
2,3
2,3

1-3
1-3
1-3
1-3
3*
3«
3«
3«
3«

NA
NA
Rabbit
1-3

1-3
1-3
1-3*
1-3^
2,3*
2,3
2,3
1-3

1-3
1-3
1-3
1
2,3
NA

NA
i-3

1-3
1-3
1-3
1-3
1-3
1-3
2,3
1
.2,3
NA
3d
3d
NA

2,3
2,3
2,3
2,3

1-3
1-3
NA
NA
NA
NA
NA
NA
NA

3f
NA
Human
S,E

S,E
NP
NP
NP
NP
NP
NP
NP

NP
NP
NP
NP
NP
NP

NP
NP

E
NA
E
E
E
E
E
NA
E
NA
Ed
Ed
NA

E
S,E
E
E

NP
NP
NP
NP
NP
NP
NP
NP
NP '

Ef
E
Reference


24-26
24-27
24,25,27-30
26, 31-34
13, 18, 35
26, 32, 33
26
32, 34

25,27
27, 36, 37
38-40
41,42
6,41,42


26,27,43
26,27,43

2, 24, 26, 44-46
24, 26, 44-46
2, 24, 26, 44-46
24, 26, 44-48
2, 24, 26, 48
2,26
2,49
2
2,42,50



2,46,51

52,53
52,53
52,53
52,53

54
54
54
54
55
55
55
55



56

-------
                                     48
                            TABLE 8 (Continued)
    "Test 1 - initial at maximum tolerated dose (MTD), or MTD and 0.7 MTD, run for exactly six
 cycles of the seminiferous epithelium. A similarly significant change (probably >15%) in any criterion
 would be evidence of an effect.
    Test 2 = dose response at MTD, at -1 and —2 log dose, and down to human level if known or until
 no response is obtained in any test; run for exactly six cycles of the seminiferous epithelium.
    Test 3 " long term, reversibility; several doses and time periods. Expose to at least three doses for
 at least 6 cycles of the seminiferous epithelium (kill 1/3 of males) and then allow recovery for 6 cycles
 (kiU 1/3 of males)  and 12 cycles (kill 1/3 of males). Recovery at  12 cycles after termination of
 treatment should be  to at least 90% of control level to show complete reversibility.
    S = procedure useful for screening humans in industrial setting.
    E = procedure useful for studying individuals thought to be exposed to an agent. Evaluation of
 human semen should use samples obtained after 2 to 5 days of abstinence with samples  taken over
 time.
    NA » not necessary.
    NP = not practical or possible.
    ^Especially important when studying recovery.
    cSave tissue from level 1 test, fix in Bouins, for possible later use.
    ''in 3 samples taken near end of treatment and then in additional samples to get clearance rate.
    ^Female rats killed 18-24 hours after mating to  evaluate fertility, sperm penetrating ability, and
 sperm transport.
    /If compound is detected in seminal plasma of  rabbit or man, incubate both rabbit sperm and
 human sperm from normal donors and determine a dose response  of sperm to the drug in vitro.
 Evaluate percentage  of motile sperm over time at 37° or in vitro penetration of hamster oocyte.
    All  of the  tests  listed  in Table  8  are feasible  in most well-
equipped  laboratories.  The  phase-contrast  microscope  and video
micrography equipment (estimated additional cost:  $6000) are the
only  nonstandard requirements.  The training period necessary to
conduct the tests in an accurate  and precise manner is not excessive.
    The  tests  selected  can be used to (a)  detect an effect of a  test
compound  on  male  reproduction  and  (b)  serve  as  a basis  for
estimating an acceptable level of exposure.
    The  tests listed will yield quantitative data that are amenable to
efficient statistical analyses and that have a sufficient range of values
to  enable establishment of dose-response curves.  The variability of
the tests is shown in Table 10 for most parameters.
    The  proposed  tests  are  for the  most part  quite  specific for
reproductive toxicity. Results of each test should not be affected by
other body  systems or, except for a possible decrease in testosterone
level, cause changes in other aspects of body function.
    Any  subchronic  or  chronic  test used  to evaluate effects of an
agent  on male  reproduction  must  extend  over  6  cycles  of  the
seminiferous  epithelium,  when  it is assumed  that an agent  bio-
accumulates to a steady state within 1  cycle (23).  This  interval is
based on (a) the time needed to reach a steady state concentration of

-------
                                          49
                 TABLE 9 Reasons for Rejection of Potential Evaluation Tests
                         Considered by Male Reproductive Subgroup
                    Test
                                            References
     Reasons for rejection"
  Tonometric measurement of testicular consistency   26,57
  Qualitative testicular histology                  24, 31, 34
  Stage of cycle at which spermiation occurs         24, 31, 34
  Quantitative testicular histology
    Counts of degenerating germ cells              18, 35,58
    Complete germ cell counts                    18, 35,58
    Stem cell counts                            18,35,58
    Relative frequency of stages of cycle            18,35,58
  Epididymal histology                         59, 60
  Biochemistry of epididymal fluids                2,61
  Histology of accessory sex glands                62
  Biochemical analysis of sperm                   2
  Sperm membrane characteristics                 63, 64
  Biochemical analysis of seminal plasma            65
  Evaluation of sperm metabolism                 65,66
  Fluorescent Y bodies in spermatozoa             67, 68
  Flow cytometry of spermatozoa                 69
  Karyotyping human sperm pronuclei             70
  Cervical mucous penetration test
  Studies on prepuberal animals
US (rat), NV (rabbit), FR (human)
I
RR,UR

UR
UR,$$
UR,$$,FR (rabbit)
UR,I
I,NR
NR
NR
NR,$$
NV.FR
NR
NR,$$
NV.NR
UR, NV, FR
FR
UR (human)
NR (rat, rabbit)6
     CUS = unsuitable for species
     NV = not validated
     RR - redundant
     I = insensitive
     NR = not relevant
     UR = only in specialist lab
     FR = future research
     $$ = too costly
     "Studies on animals treated prior to puberty have not been included for the following reasons. It
  would involve a redetermination of maximum-tolerated-dose levels for young, growing animals. The
  choice of age period of exposure is a complex topic and sufficient time was not available to adequately
  consider  this. Humans are exposed to many of the agents that would cause reproductive problems
  primarily through occupational exposure after puberty. Some agents (radiation, cyclophosphamide)
  that cause reproductive problems with prepubeial exposure also affect postpuberal males. Nonetheless,
  unique developmental processes occur in testicular development prior to and during puberty, and
  therefore a possibility exists that some agents would only affect the prepuberal male. The group of
  tests proposed in Table 8  would provide a sensitive measure of such effects, if animals exposed at any
  time during puberty or throughout their development were analyzed after reaching sexual maturity.
agent in the target organs of the rabbit or rat, (b) the concepts that
an agent acting directly or indirectly on the germinal epithelium may
act on  a  specific type  of  cell and that affected germ  cells  may
develop  for some time  before  they  degenerate, (c)  the fact  that
damage to germ cells is most  evident by absence of certain types of
germ  cells,  and  (d)  qualitative change  in germ  cells  may  not  be
readily  discernible  until active  spermatozoa  pass  into  the cauda
epididymidis or ejaculated semen. The present protocol assumes that
attainment of a steady state  concentration of an agent requires an
interval equal  to  one cycle  of the  seminiferous epithelium.  Forma-
tion of primary spermatocytes from renewing spermatogonia in the

-------
50
TABLE 10 Approximate Variation Between Animals for Suggested Test
Criteria (CV)fl Coefficient of Variation (%)
Rabbit model0

Criterion
Body weight
Testis
Weight
Size in situ
Spermatid reserves per testis
Spermatid reserves per gram
Tubule diameter
Epididymis
Weight of distal half
Number of sperm in distal half
Motility of sperm, distal end (%)
Gross sperm morphology, distal end (%)
Detailed sperm morphology, distal end (%)
Accessory sex glands
Weight vesicular glands
Weight total accessory sex glands
Semen
Total volume
Gel-free volume
Sperm concentration
Total sperm per ejaculate
Total sperm per day of abstinence
Sperm motility, visual (%)
Sperm motility, videotape (%)
Gross sperm morphology
Detailed sperm morphology
Concentration of agent in seminal plasma
Concentration of agent in blood
Endocrine
Luteinizing hormone
Follicle-stimulating hormone
Testosterone
Gonadotrophin-releasing-hormone stimulation
Fertility^
Ratio of exposed to pregnant females
No. embryos or young per exposed female
No. embryos or young per pregnant female
Ratio of embryos to corpora lutea
Rat model6
(Wistar)
20

5
NAa
11
8
_e

13
20
-
-
-

26


NA
NA
NA
NA
NA
NA
NA
NA
NA
NA
NA

80
65
33
-





"Data are not available to allow calculation of sensitivity of the
*Ref. 27.
cRefs.25,37,and44.
j j '
NA = not applicable.
e— = data not available.
•fipor controls: with treatment, variability may




be greater.
(New Zealand
White)
37

18
9
24
9
5

13
52
-
-
-

10
-------
                                51

rat requires about 1.5 cycles, and spermiation occurs about 3 cycles
of the seminiferous epithelium after those sperm have  become
primary  spermatocytes.  Passage  of sperm through the epididymis
into  the  distal cauda or ejaculated semen requires 1.0 to 1.5 cycles,
depending on  the  species and  frequency  of  ejaculation.  Conse-
quently,  if an agent acted on A-type spermatogonia, a decrease in
number  of  sperm ejaculated or in the fertility of sperm from the
cauda  epididymidis  might  not  occur   for   5  to  6  cycles
(1.5  + 3.0 + 1.0 = 5.5 cycles) of the seminiferous epithelium. If the
agent  resulted in  degeneration  of  pachytene  spermatocytes,  an
alteration in semen  characteristics or fertility might be expected to
occur after 4.0 to 4.5 cycles. However, with  continuous exposure to
the  test  compound, such a lesion would remain detectable in the
semen  or by examination of testicular histology at the end  of 6
cycles of the seminiferous epithelium.
   By testing male rats or rabbits for their fertility after 5 cycles, a
depression in fertility caused by  a compound inducing  a qualitative
change in sperm  function should be detectable, since this probably
would  affect spermatocytes or spermatids. Allowing 6  to 8 days of
sexual  rest between the end of fertility testing and necropsy of test
males after 6 cycles provides time for restoration  of the  normal
population of sperm in the cauda epididymidis hi males receiving
doses  that  do  not suppress daily  sperm  production. If sperm
production is low in test males,  the reserve level in the cauda will
reflect this,, but sufficient sperm may still be  present to allow
assessment of sperm motility and morphology.
   For these reasons, evaluation of an agent, administered chroni-
cally, for effects  on male reproduction should include fertility  tests
after 5 cycles and examination of the testes, epididymides, accessory
sex  glands,  and plasma hormone levels after  6  cycles   of the
seminiferous epithelium.
   If  an agent  is  shown to  alter  male reproduction  in a test
extending over 6 cycles  of chronic  exposure (Test 1  or Test 2 as
described under  protocols for  testing),  it  may be  desirable  to
determine if the  effect is reversible (Test 3). A test of reversibility
should extend over 18 cycles of the seminiferous epithelium.  Chronic
exposure to  the agent should extend  over 6 cycles, and 12 cycles
should elapse  after  the termination  of  exposure to allow for
restoration of normal reproductive function.
   Although the recovery  period  in  man is usually longer, these
animal data should provide a clear indication if complete recovery
will  occur in man (compare radiation data of Meistrich et al. [13] in
the mouse and Rowley et al. [ 14]  in man).

-------
                               52

        Evaluation of Reproductive Damage in Exposed or
                    Potentially Exposed Men

General
   Two types of studies in humans seem particularly relevant to the
objectives of a male reproduction risk assessment. The first involves
surveillance studies, in which periodic checking is done on men in a
setting (e.g., industrial  or  agricultural)  that might, in the future,
involve the  risk of a reproductive defect. An example  of such a
setting would  be a chemical  company in which substances  are
prepared that are known from  animal studies to cause reproductive
toxicity when administered in high dosage but not at a dosage up to
10 times that expected  for human exposure. The safety factor may
be variable, however, and depends on the quality of the animal study
from which it was derived. This type of surveillance is important for
at least two reasons: (a) the sensitivity of men may be greater than
allowed for  by  the tenfold safety factor and (b) the exposure of the
workers may be greater than originally  estimated. The methods for
surveillance  of this type could be quite  innocuous and could be
incorporated into an annual medical checkup if this were already a
practice.
    The second type of study would be of men who have been or are
being exposed to a known reproductive toxin in dosages likely to be
toxic in man as based on animal studies. This type of study could be
used in men exposed to high dosages of one or more general toxins
for  which the  effect on testicular function had not been studied
carefully in animals.

Surveillance studies
    Men  could be asked yearly whether they have been attempting to
cause a pregnancy and have been unable to do so. The prevalence of
infertility in couples within the reproductive age group  is approxi-
mately 15% (74). If more than 20% of men between 19 and 35 have
been unable to produce a pregnancy in over one year of unprotected
intercourse, a possible toxic effect should be looked for in a rigorous
manner  as  outlined  in the  following section  on  known  toxic
exposure.
    Testicular length could  be  measured on annual physical exami-
nation. If the distribution of testicular size  for men falls significantly
below the lower norm (3.5 cm  for Caucasian and Black) for that age
group and  ethnic  background  (76),  a  toxic  effect  should  be
suspected.

-------
                             .53

    Blood levels of follicle-stimulating hormone could be measured
yearly. If mean levels significantly vary from those of age-matched
controls, a toxic effect should be suspected.
    If any of these three variables suggest testicular toxicity, a more
detailed study  of the population should be undertaken, as outlined
below.

Study of men with known toxic exposure

    Where a human population is suspected of being at reproductive
risk because of environmental hazards, a number of potentially toxic
agents may be involved, and the duration and level of exposure may
vary within the population. Although each potential toxicant should
be  carefully tested by the laboratory screening methods outlined in
this document, it would be useful to make a more immediate and
direct assessment of fertility  potential in the exposed population.
The requirements for this include capacity for rapid response to the
subjects, feasibility,  sensitivity,  and  data that  can  be analyzed
statistically. The data obtained in such studies should provide an
initial indication of  the degree of testicular damage,  and where an
environmental reproductive hazard has been identified, more detailed
studies may be undertaken to characterize objectively male reproduc-
tive dysfunction.
    To carry out these studies, a  specialized team and  a  modest
amount of equipment would be needed. The latter could be installed
at  locally  available facilities, or  a mobile  laboratory  could be
equipped. Detailed medical, reproductive,  and occupational histories
should be taken from each exposed subject and a physical examina-
tion given.  At least five semen samples should  be  evaluated per
individual at two-day intervals. Objective data on testicular size and
consistency could be obtained by sonography and tonometry. Blood
and urine could be obtained at this time for endocrine studies and/or
toxicant levels. Controls to be studied must be carefully chosen and
matched.  Before a  national  data  base is established, individuals
should be  selected  according to epidemiological advice. For the
details  of these analyses,  see "Study of men with known  toxic
exposure" in the Appendix to this chapter.
    Statistically significant differences between the exposed and the
control groups  (matched for age, occupation, geographical location)
in  seminal  fluid  and  blood hormone  measurements  would be
evidence for  an effect  of the exposure on  male  reproductive
function. An adverse effect would be expected to decrease sperm
counts, motilities, and numbers of sperm with normal morphology; if

-------
                                54

the effect were sufficiently severe, blood testosterone levels would
decrease. If the toxic effects were directly on the testis (as is the case
with  the  great  majority of  known  toxins), follicle-stimulating
hormone (FSH) levels would increase. With a mild  toxic effect on
the testis,  blood FSH levels after administration  of gonadotropin-
releasing hormone  (GnRH) might exceed  normal responses,  even
when basal FSH levels are normal. If the toxic effect were primarily
on the pituitary gland or central nervous system, luteinizing hormone
(LH) and FSH levels would tend to decrease.
    If  not  established initially (Surveillance studies, v.  sup.), other
comparisons between the exposed and control groups should include
(a) rate of infertility as indicated by the number  of men who have
not been able to  induce a pregnancy in over one year of intercourse
without  using contraceptives, and (b) testicular size, with particular
attention to  the number of men with testicular length less than
3.5 cm.
    Differences between the exposed and control groups in these last
two assessments, suggested also for  the initial screen, will be found
when reproductive  toxicity -is sufficiently severe. However, measure-
ments of fertility and testicular  size would be  expected to be less
sensitive in revealing mild defects in  gonadal function than the
seminal fluid and blood hormone measurements described above.

Additional comment on human testing procedures
    Blood  samples  for  hormone   measurement  and  noninvasive
procedures  such as testicular  length  may be the more  feasible
parameters to evaluate because of the added difficulty in obtaining
semen samples, in some human populations at least. However, where
these give equivocal results, it is likely that semen analyses will help
to resolve the fundamental dilemma.
    An elevated FSH  level is  a sensitive indicator  of decreased
function  of the germinal epithelium  in  man  and  experimental
animals. However, while there is no doubt that increased FSH levels
usually  imply decreased sperm production (9,  14,  21,  77, 78),
measuring FSH levels is probably a less dependable test than direct
sperm counts, for  it  is a consistent  marker  only of severe oli-
gozoospermia or azoospermia (53,  79,  80). Measurements of FSH
seem useful  adjuncts to  sperm  counts,  therefore, and indicators of
the direct action of toxic agents on pituitary function. Despite their
wide variability in man, total sperm per ejaculate  are usually a more
sensitive measure of testicular damage than elevated FSH levels.

-------
                                55

    Comprehensive semen analysis requires an assessment of sperm
morphology as well as total sperm per ejaculate. This aspect of the
human  ejaculate  has  received considerable general  comment as a
parameter that also often  falls below the  standards that might be
expected for animals living in the same area. Since about 30% of the
spermatozoa are abnormal in semen from a presumed  fertile group of
men, only by using large groups of 100 or so persons would it be
possible to detect increases in abnormal spermatozoa of the order of
10% in cross-sectional studies in which only one to four samples are
collected for each man in  exposed and control groups. Despite the
relatively variable morphology of a significant proportion of sperma-
tozoa in the human ejaculate, sperm morphology tends to be fairly
constant for one individual (81). This justifies the use of fewer men
in longitudinal studies  where such studies can be  undertaken (as
compared to postexposure analyses),  since the men can then act as
their own controls (81).
    Human  sperm morphology  classification is currently subjective,
personality oriented, and nonstandard (82). However, detailed "type
classification" (i.e., oval versus tapering versus amorphous) may not
be required  to identify groups of individuals at reproductive risk. In
normal  fertile human  semen, sperm  morphology is relatively uni-
form, more  than 50% of the sperm  having the typical "oval" shape.
In contrast, infertile human semen is characterized by a diversity of
abnormal sperm sizes and  shapes. If  objective, morphometric data
describing sperm  size and shape (e.g., head length, width, area, and
circumference; tail midpiece width) were obtained from individuals
at potential risk, these could  be compared statistically  with data
from the matched control group. Significantly greater dispersion in
the morphometric parameters of the  exposed group might indicate
increased reproductive dysfunction in the population.  The magnitude
of  differences between the exposed  group and  the control group
might also provide an indication of the severity of testicular damage.
As  noted earlier, the methods  for automatic evaluation of sperm
morphology are not well established  and  need considerable  refine-
ment and validation (6).

Assessment of risk to men

    Assessment of risk to reproductive performance  and fertility in
men is inadequately tested at present. The quantitative assessment of
risk to general human health from exposure  to  environmental
toxicants has been approached by relating the probable or estimated
dose of a suspect toxic agent to the occurrence of deleterious effects

-------
                                56

on the basis of either epidemiologic data on human populations or of
experimental data from animal studies. It seems likely that threshold
effects will appear for most agents. Ideally, assessment of the effects
of these agents upon male reproductive function should be based
upon human epidemiologic data. However, there are few epidemio-
logic  risk assessment  data regarding  the  effect of environmental
agents on the fertility of men. Thus,  at present a quantitative risk
assessment must  depend on extrapolation  to man of measurements
of  the reproductive  end  points  in  experimental   animal  systems
discussed here.  One approach to  risk assessment estimates  the
acceptable daily intake (ADI) of a chemical, defined as the exposure
level that is anticipated to be without risk to the species. It should be
cautioned that the ADI  represents  only a judgment, is  not an
estimate of  risk nor a guarantee of absolute safety, and is subject to
modification as additional relevant information becomes available.
    To  account for the uncertainties involved in extrapolating from
animals to man, the ADI includes an uncertainty or safety factor to
the highest  no-adverse-effect level measured in an  animal study. A
no-adverse-effect level  is  defined here as  a dose  for  which no
significant difference is found between control and treated animals
for any of the end points measured adequately. It is important that a
statistically  significant  effect also be biologically  significant.  This
uncertainly factor  will depend on (a) the animal species/strain; (b)
the  quality  of  the  experimental  data;  (c)  the  availability  of
comparative pharmacokinetic information on the animal species' and
man's absorption, distribution, metabolism, binding, and elimination
of the chemical;  and (d) any other relevant comparative information
on structurally similar chemicals. In the absence of these comparative
data, we should follow the guidelines of the Safe Drinking Water
Committee, National Research Council of  the National Academy of
Sciences (83), and recommend an  arbitrary uncertainty factor of 100
for adequate animal studies. In the case of human male reproduction,
the  size  of this factor seems more than justified by increasing
evidence that the human testis functions less efficiently and possibly
closer  to a point of  pathology than that of the animal models
recommended (2, 7, 8). Thus, for  an agent  causing a reversible action
in  a model animal,  the ADI would be 0.01 times the no-adverse-
effect level for the most  sensitive criterion and the most  sensitive
species evaluated, whether rat or  rabbit. A daily exposure or intake
above this  level  represents a risk  of reproductive damage to human
males. For  irreversible effects on male reproductive function, we feel
we can make no recommendation for a quantitative risk assessment.

-------
                                57

       Protocols for Testing Compounds with Animal Models

    The actual criteria to be evaluated in each test are shown in Table
8. The time schedules for conducting Tests 1, 2, and 3 are shown in
Table 11. If pharmacological  studies show that the test compound
may bioaccumulate so that the body  burden increases beyond an
interval equal  to one  cycle  of the  seminiferous  epithelium, the
treatment  interval  of both Test 1  and Test 2 must  be  increased
appropriately.  At  least  5  cycles  should  elapse  after  reaching
maximum body load.

Test 1 — initial screen

    As an initial screening procedure, animal exposure will be greater
than or equal to half of a maximum tolerated dose (>0.5 MTD) of
the test agent for an interval  equal to  6 cycles  of the  germinal
epithelium. An initial screen using an acute exposure is considered to
be unnecessary, because the subchronic test is more sensitive.
    To initially assess risk to male reproduction, a compound should
be  subjected  to  in vivo tests utilizing both rats  and rabbits. A
compound producing no statistically significant alteration in  any
criterion for either species  when  given at  >0.5 MTD would  be
considered to  be  safe  for  humans  (see safety  factor in  risk
assessment). A statistically significant alteration in  any criterion
would necessitate  conduct of  a Test-2 evaluation to establish a
dose-response curve, unless manufacture or  use of the agent were to
cease, or if a larger safety factor were used. Test 1 (Tables 8 and 11)
uses both rats and rabbits and  is detailed in the Appendix to this
chapter.  The  fixed  time  schedule is  designed to  maximize the
probability of detecting any decrease in reproductive function.

Test 2 — dose response curve

    1. The general approach used in the initial screen (Test  1) will be
used except  that additional  criteria of reproductive damage are
included (Table 7).  The dose-response curve will include at  least
three points, usually the dose used in  Test 1, and  —1 and —2 log
doses and must extend down to the human exposure level (if known)
or until no statistically significant response is obtained in any test.
Both  0-dose  and untreated controls could be  included to detect
effects of handling that  might be associated with agent administra-
tion. If necessary,  additional  tests will be  run  to attain these  end
points. Both rats  and rabbits must be used. The fixed time schedule
(Table 11)  is essential to measure accurately the extent of damage to

-------
                                      58
          TABLE 11  Chronology of Conduct for Test with Animal Models
                          (Expressed as Day of Study)	
                      Test
                                                  Rat
                                                             Rabbit
     Test 1 or 2a
       Condition males
       Obtain preexperimental body weight
       Evaluate preexperimental semen
       Initiate compound administration
       Continue compound administration
       Weigh weekly
       Collect experimental semen (each 3-4 days)
       Measure testis size weekly
       Expose to females or artifically
         inseminate females
       Sexually rest males
       Kill males
       Kill females
       Allow females to kindle
     Test la
       Condition males
       Obtain preexperimental body weight
       Evaluate preexperimental semen
       Initiate compound administration
       Continue compound administration
       Collect experimental semen
       Expose to females or artifically
         inseminate females
       Kill 1/3 of males
           1/3 of males
           1/3 of males
       Kill females 18—24 hours past mating
                 12—18 days past mating
       Allow females to kindle
-21-0
-14 + 0
NA6
day 0
0-77
0-78
NA
NA
65-71

71-78
78
83 - 89C
NA

-21-0
-7 + 0
NA
0
0-77
NA
NA
NA
215-221

78
155
232
216-222
233
NA
-28-0
-14 + 0
-14-0
day 0
0-64
0-65
3-4-53-54
0-65
54-57

58-65
65
NA
85-90

-28-0
-7 + 0
-14-0
0
0-64
35-64
118- 140
158-194
184-187

64
128
193
NA
NA
215-218
         "Rats will be weighed weekly. Rabbits will be weighed weekly, testis size
      measured weekly, and semen will be collected twice weekly (every 3 to 4 days).
      Schedule is for a compound that does not accumulate for a long time; steady
      state level in body tissues reached in <10—12 days.
         °NA = not applicable.
         cKill females 18 days after mating, as determined by a vaginal smear.
the  different aspects of male reproductive function and to enable a
prediction of human risk.
    2. In evaluating testicular histology, sections representing at least
two loci will be used. The diameter of 50 tubules will be measured;
the percentage of seminiferous tubule cross-sections (N = 250) having
mature spermatids lining the tubule lumen and the percentage  of

-------
                                 59

 tubules (N = 250) devoid of germ cells other than spermatogonia will
 be determined. Evaluations of the morphology of sperm in the cauda
 epididymidis and ejaculated semen will be more comprehensive than
 in Test 1. The serum concentrations of luteinizing hormone (LH),
 FSH, and testosterone also will be determined.

 Test 3 —  recovery study

     1. Test 3 is a long-term study designed to test the reversibility of
 damage to male reproduction and also to evaluate sperm transport,
 penetration of sperm into ova, and early  embryonic death. Many
 agents  that  cause  degeneration of the germinal epithelium  and
 azoospermia will not damage the stem spermatogonia. If the latter
 remain, eventual recovery of the germinal epithelium is likely. This
 test measures recovery of the germinal epithelium and fertility, at 6
 cycles and 12 cycles (155  days for rats  and  128 days for rabbits)
 after ending a 6-cycle exposure to the  test compound. Although
 recovery  of the germinal epithelium might  not be complete by 12
 cycles  after exposure,  some onset of recovery probably should be
 detectable by then if it will occur eventually. If the test compound is
 one known to  bioaccumulate, longer treatment periods (as used in
 Test 2) and  recovery periods (at least twice  the  duration  of the
 treatment period) are essential.
    2. The criteria evaluated (Tables 8 and 11) are the same as those
in Test 2, except  that data on fertilized  rat eggs are necessary.
Consequently,  each male  will be exposed to four female rats. Two
females will be killed 18 to 24 hours after mating (as determined by
the presence of vaginal plugs) and ova recovered by flushing. The two
other  female  rats  will  be killed  12  to  18 days after mating.
Measurement  of concentrations of the compound in blood and
seminal plasma at steady state are desirable,  since these data may be
useful in  predicting potential  damage in humans and the prognosis
for recovery from such damage.
    3. The time  schedule (Table  11) for conduct of the study could
be modified by extending the treatment beyond 6 cycles, but the
timing of evaluations between days  215 and 233 for rats and days
184 and 218 for rabbits may not need to be altered.
   4. Complete reversibility is  considered to  be restoration, to at
least 90%  of control levels  at 12 cycles after cessation of exposure, of
all criteria adversely altered in males after 6 cycles of exposure (in
Test 2 or Test 3) at a given dose.

-------
                               60

                        Research Needed

   Until about 15 years ago only outline information  about the
male tract was available  (84—86).  Although much precise data has
appeared since then for the animal models suggested and  even some
for man (65, 87, 88), it is difficult to  compare the two. Research
into this and related aspects as suggested below is a critical element
for establishment of a reliable assay and evaluation of risk in males.
    1. Variance  components  for  characteristics  of  semen (total
volume, total sperm per ejaculate, percentage of motile sperm, and
incidence of sperm  abnormalities) are  available  for rabbits   (see
Table 10) but have  not  been reported for man. This information
should be obtained for men  of different age groups (<20, 21-30,
31_40, >40)  with different life styles or occupations,  so  that
efficient and meaningful evaluations can be made.
   2. The influence of an abstinence interval on characteristics of
human semen should be  evaluated  critically for men of 20 to 30, 30
to 40, and >40 years of age. Procedures for reducing the influence of
an  abstinence  interval  on  estimates  of sperm  production  (e.g.,
normalization of data for each  ejaculate by dividing by the number
of days of abstinence) should be  evaluated.  It is also unclear  what
effect repeated ejaculation has on the absolute  concentrations of
many seminal compounds that might be measured as indicators of
the activity of accessory glands in man  or the amount of the test
agent in seminal plasma.
    3. For the human, the relationship should be determined among
testicular size, tonometric measurements or testicular consistency,
and sperm output as well as other ejaculate characteristics.
    4. Relationships  need to  be established among testicular histol-
ogy, ejaculate  characteristics, sperm morphology, and fertility of
humans, rabbits, and rats. Indices of fertility should be calculated.
    5. Automatic or semiautomatic morphometric procedures should
be further  developed for analysis of  the  morphology  of human
spermatozoa and spermatozoa from  test animals. First-generation
systems for automated evaluations are  available (e.g., at Lawrence
Livermore Laboratory),  but  the instrumentation and software need
additional refinement and validation before these techniques can be
applied routinely in analyses of human sperm morphology (89, 90).
    Sperm morphometry can be obtained for living sperm cells or
from stained seminal smears,  by methods  becoming increasingly
automated, either using, flow cytometry (89) or tracing sperm shape
from the screen of a video monitor using an electronic planimeter-
digitizer integrated  into a minicomputer (49). The  use of sperm

-------
                               61

morphology for diagnosis or prognostication of specific reproductive
disorders  will require a "type classification" of individual  sperm
abnormalities. The determination of such  a classification can be
made by computer on the basis of the morphometric data obtained.
Research should be encouraged to develop such computer software.
A classification system based on morphometric standards should be
developed.
    6. A  data bank should  be established for (a)  the control data
from screening tests with  animals  to build a  large  base for
computation of variations associated with each characteristic among
trials,  locations,  season,  year,  etc.; (b) the  chemical nature of
compounds tested and found in an initial screen  to  damage some
aspect of  male reproduction or to have no effect; and (c) the
chemical nature of all compounds found to have a deleterious effect
on male reproduction, the nature, extent, and incidence of damage in
each exposure dose, and interval to recovery.
    7. Available data on effects of agents known to alter human male
reproduction should be correlated with data on their action  in test
animals in a battery of tests. The repeatability and relative sensitivity
of the  tests within  and between  species  should  be  determined.
Recommendations for specific studies are as follows:
    (a) Obtain more extensive and accurate analytical data on semen
from men exposed to chemotherapy and the effects of parallel levels
of chemotherapy in animals.
    (b) Bring  together  existing  radiation  studies  in human  and
experimental animals for development  of models in which to base
chemical risk assessment data.
    (c) Obtain  better  data  in  experimental  animals  on effects of
dibromochloropropane or other agents known to be harmful to man.
    8. The relative usefulness of basal FSH concentration in blood or
of FSH response to GnRH as indicators of testicular damage should
be compared with seminal analyses to determine their sensitivity (see
Table 10  and Appendix to this chapter).  For screening of large
numbers of human males, it would be useful to know the single most
sensitive index of testicular toxicity. In man, a test based on a blood
sample is more practical than one requiring submission of a seminal
fluid sample.
    9. The  responsiveness  of  Leydig  cells  should  be evaluated.
Well-characterized in vitro  bioassays for LH have  been developed.
Rat or mouse Leydig cells  are incubated over several hours with
various concentrations of LH. The amount of testosterone produced
by the Leydig cells is measured. A potential testicular toxin could be
studied by exposing it,  in various  concentrations,  with LH  to the

-------
                                62

Leydig cell preparations and comparing the amount of testosterone
produced to that produced by cells exposed to LH alone.
    10. The responsiveness  of Sertoli  cells should  be evaluated.
Sertoli cell cultures, preferably from postpubertal males, can be used.
The production of known secretory products, such as transferrin or
androgen-binding protein,  following  stimulation  by FSH,  can be
measured as an index of their activity. An effect of a toxin would be
reflected in that index. A major question is the relationship of
in vitro  toxicity to in vivo  toxicity, particularly  considering the
short-term  nature of the tests and the  long-term  nature of in vivo
exposure.  However,  some  of the in vitro  tests  provide  limited
opportunity to evaluate human tissue directly with animal models.
    11. Competitive mating (heterospermic insemination) should be
evaluated as a screening assay. The use of a mixed-insemination assay
(91—93) for screening  toxicants offers  a means  of increasing the
sensitivity of fertilization assays and should be explored. Rabbits and
possibly rats could be used. Semen from exposed and control males
would be  mixed and inseminated into the same female  and the
paternity of the offspring established by genetic markers (i.e., eye or
coat color). This has good potential for use as a screening assay of
superior sensitivity which could simultaneously assess disturbances of
sexual behavior, sperm quality,  sperm  transport in the  female,
fertilization,  and embryonic and fetal  development. A  limited
number of trials with the system should be adequate to determine its
utility.
    12. The direct assessment of damage to the sperm genome would
permit routine screening and monitoring of males for exposure in the
workplace  to  chemicals that may be  hazards to their reproductive
capacity. Further studies might attempt the following:
    (a) Establish the degree of correlation between abnormal sperm
head morphology and an aberrant chromosome complement.
    (b) Develop sensitive methods for the identification and measure-
ment of alkylated or modified DNA bases.
    (c) Improve  the  methodology to quantitate  alkylated  amino
acids, since there is  evidence that alkylation  of  sperm chromatin
proteins also contributes to reproduction failure.
    (d) Develop methods to detect damage to sperm chromatin (e.g.,
enzymatic detection of strand breaks in sperm DNA).
    Additional studies could be designed  and sponsored to evaluate
the  suitability of the four techniques  discussed  above as  routine
procedures for detection of genetic abnormalities by direct observa-
tion of spermatozoa and of the male pronucleus.

-------
                                   63
                            REFERENCES

 1. Smith, K. D., Rodriguez-Rigau, L. J. and Steinberger, E.:  Relation between
     indices of semen analysis and pregnancy rate in infertile couples. Fertil.
     Steril.28: 1314-1319,1977.
 2. Amann, R. P.:  A  critical review of methods for evaluation of spermato-
     genesis from seminal characteristics. J. Androl. 2: 39—60, 1981.
 3. Barfield, A., Melo, J., Coutinho, E., Alvarezs, P., Faundes, A., Brache, V.,
     Leon, P., Frick, J., Bartsch, G. and Weiske,W. H.: Pregnancies associated
     with sperm concentrations below 10 million-mi in clinical  studies of a
     potential male contraceptive method,  monthly depot medroxyproges-
     terone acetate and  testosterone esters. Contraception  20(2):  121—127,
     1979.
 4. David, G., Jouannet, P., Martin-Boyee,  A., Spira,  A. and Schwartz, D.:
     Sperm counts in fertile and infertile  men. Fertil. Steril. 31(4): 453—455,
     1979.
 S.Zuckerman, Z., Rodriguez-Rigau, L. J., Weiss, D. B., Chowdhury, A. K.,
     Smith, K. D. and Steinberger, E.: Quantitative analysis of the seminiferous
     epithelium in- human testicular biopsies and the  relation of spermato-
     genesis to sperm density. Fertil. Steril. 30: 448-455, 1978.
 6. Freund, M.: Standards for the rating of human sperm morphology. Int. J.
     Fertil. 11:97-180,1966.
 7. Johnson, L., Petty, C. S. and Neaves,  W. B.: A comparative study of daily
     sperm production and testicular composition in humans  and rats. Biol.
     Reprod. 22: 1233-1243,1980.
 8. Amann, R. P.  and Howards,  S. S.:  Daily spermatozoal production and
     epididymal spermatozoal  reserves of  the human male.  J.  Urol.  124:
     211-215,1980.
 9. Smith, K. D. and Steinberger,  E.: What is oligospermia? In: The Testis in
     Normal and  Infertile Men, D. Troen and H. Nankin, Eds., Raven Press:
     New York; pp. 489-503, 1977.
10. Salisbury, G. W.,  Van Demark, N. L.  and Lodge, L. R.:  Physiology of
     reproduction  and artificial insemination of cattle, W. H.  Freeman and Co.:
     San Francisco; 788 pp., 1978.
11. MacLeod, J., Hotchkiss,  R. S.  and Sitterson, B. W.: Recovery  of male
     fertility after sterilization  by nuclear radiation. J. Am.  Med. Assoc. 187:
     637-641,1964.
12. Amelar, R. D., Dubin, L. and Hotchkiss, R. S.: Restoration of fertility
     following unilateral orchiectomy and  radiation therapy for testicular
     tumors. J. Urol. 106: 714-718, 1971.
13. Meistrich, M. L., Hunter, N., Suzuki, N., Trostle, P. K. and Withers, H. R.:
     Gradual  regeneration  of  mouse  testicular  stem  cells  after  ionizing
     radiation. Radiat. Res. 74:  349-362, 1978.
14. Rowley, M. J., Leach, D. R.,  Warner, G. A. and Heller, C. G.: Effect of
     graded doses of ionizing  radiation  on human testes.  Radiat.  Res. 59:
     665-678, 1974.

-------
                                   64

IS.Cheviakoff, S.,  Calamera,  J.  C., Morgenfield, M. and  Mancini,  R. E.:
     Spermatogenesis in patients  with  lymphoma after treatment  with  chlo-
     rambucil.J.Reprod. Fertil. 33: 155-157,1973.
16. Sherins, R. J. and DeVita, V. T., Jr.: Effect of drug treatment for  lymphoma
     on male reproductive capacity—studies of men in remission after therapy.
     Ann. Intern.Med. 79: 216-220,1973.
17. Sieber, S. M. and Adamson, R. H.: Toxicity of antineoplastic agents in  man:
     chromosomal aberrations, antifertility effects, congenital malformations,
     and  carcinogenic potential. Adv. Cancer Res. 22: 57—155,1975.
18. Lu, C. C. and Meistrich, M. L.: Cytotoxic effects of chemotherapeutic  drugs
     on mouse testis cells. Cancer Res. 39: 3575-3582,1979.
19.Torkelson, T. R., Sadele, S. E., Rowe, V. K., Kodama, J. K., Anderson,
     H.H., Loquvam,  G.S. and Hine, C.H.:  Toxicologic investigations of
      l,2-dibromo-3-chloropropane. Toxicol. Appl. Pharmacol. 3:  545-559,
      1961.
20. Whorton, M. D., Krauss, R. M., Marshall, S. and Milby, T. H.: Infertility in
     male pesticide workers. Lancet 2(8051): 1259-1261,1977.
21. Glass,  R. I., Lyness, R. N., Mengle, D. C., Powell, K. E. and Kahn, E.: Sperm
     count depression in pesticide applicators exposed to dibromochloropro-
     pane. Am. J.Epidemiol. 109(3): 346-351,1979.
22. Lantz, G. D., Cunningham, G. R., Huckins, C. and Lipshultz, L. I.: Recovery
     from severe oligospermia after exposure to dibromochloropropane. Fertil.
     Steril. 35: 46-53,1981.
23.Amann, R. P.: Use of animal models for detecting specific alterations in
     reproduction. Appl. Fund. Toxicol.; in press, 1982.
24. Amann, R. P.: Sperm production rates. In: The Testis, Vol. 1, A. Johnson,
     W. Gomes, and  N. Van Demark, Eds., Academic Press, Inc.: New York;
     pp. 433-482,1970.
25. Carson, W. S. and Amann, R. P.: The male rabbit. VI. Effects of ejaculation
     and season on  testicular  size and  function. J. Anim. Sci. 32: 302—309,
      1972.
26. Foote, R. H.: Research techniques to study reproductive physiology in the
     male.  In: Techniques and  Procedures in Animal Science  Research.
     American Society of Animal Producers: Albany, New York; pp. 81 — 100,
      1969.
27. Robb, G. W., Amann, R. P. and Killian, G. J.: Daily sperm production and
      epididymal sperm reserves of pubertal and adult rats. J. Reprod. Fertil. 54:
      103-107,1978.
28. Amann, R. P. and Lambiase, J. T.: The male rabbit. III. Determination of
      daily sperm production by means  of testicular homogenates. J. Anim. Sci.
      28:  369-374,1969.
29. Amann, R. P., Kavanaugh, J. F., Griel, L. C., Jr. and Voglmayr, J. K.: Sperm
      production of Holstein  bulls determined from   testicular spermatid
      reserves,  after  cannulation of rete testis or vas deferens,  and by daily
      ejaculation. J. Dairy Sci. 57: 93-99,1974.

-------
                                    65

30t Johnson,  L.,  Petty,  C.  S.  and  Neaves,  W. B.:  A  new  approach to
     quantification of spermatogenesis  and its  application to  germinal cell
     attrition during human spermiogenesis. Biol. Reprod. 25: 217—226, 1981.
Sl.Clermont,  Y.: Kinetics of  spermatogenesis in  mammals:  seminiferous
     epithelial cycles and spermatogonial renewal. Physiol. Rev. 52: 198—236,
     1972.
32. Swierstra, E. E. and Foote, R. H.: Cytology and kinetics of spermatogenesis
     in the rabbit. J. Reprod. Fertil. 5: 309-322,  1963.
33. Swierstra, E. E., Whitefield, J. W. and Foote, R. H.: Action of amphotericin
     B (fungizone)  on spermatogenesis in the rabbit.  J.  Reprod. Fertil. 7:
     13-19,1964.
34. Berndtson, W. E.:- Methods for quantifying mammalian spermatogenesis: a
     review. J. Anim. Sci. 44: 819-833,  1977.
35. Lu, C. C., Meistrich, M. L. and Thames, H. D.: Survival  of mouse testicular
     stem cells after gamma or neutron irradiation. Radiat. Res. 81: 402—415,
     1980.
36. Amann, R. P.: The male rabbit. IV. Quantitative testicular histology and
     comparisons between daily  sperm production as determined histologically
     and daily sperm output. Fertil. Steril. 21:  662-672,1970.
37. Lambiase, J. T., Jr. and Amann, R. P.: The male rabbit. V. Changes in sperm
     reserves and resoiption rate  induced by ejaculation  and sexual rest. J.
     Anim. Sci. 28: 542-549, 1969.
38. Fray, C. S., Hoffes, A.  P. and  Fawcett, D. W.: Reexamination of motility
     patterns of rat  epididymal spermatozoa. Anat. Rec. 173: 301—308, 1972.
39. Gaddum, P.: Sperm maturation in male  reproductive tract—development of
     motility. Anat. Rec. 161: 471-482, 1968.
40. Blandau, R. J. and Rumery, R. E.:  Relationship of swimming movements of
     epididymal  spermatozoa to their fertilizing capacity. Fertil. Steril. 15:
     571-579,1964.
41. Lock, L. F. and Scares, E. R.: Increases in morphologically abnormal sperm
     in rats exposed to 60Co irradiation. Environ. Mutagen. 2: 125-131, 1980.
42. Harasymowyez, J., Ball, L. and Seidel, G. E., Jr.:  Evaluation of bovine
     spermatozoal morphologic features  after staining or fixation. Am. J.  Vet.
     Res. 37: 1053-1057,1976.
43. Holtz, W.  and Foote, R. H.: Anatomy of reproductive system in male Dutch
     rabbits (Oryctolagus cuniculum)  with special emphasis on the accessory
     sex glands. J. Morph. 158: 1-20,  1978.
44. Amann,  R. P.:  Effect  of  ejaculation frequency  and breed on semen
     characteristics  and  sperm  output of rabbits.  J. Reprod.  Fertil.  11:
     291-293,1966.
45. Holtz, W. and Foote, R. H.: Sperm production, output,  and urinary loss in
     the rabbit. Proc. Soc. Exp. Biol. Med. 141: 958-962,1972.
46.Kirton, K.  E., Desjardins, C.  and Hafs,  H. D.:  Levels of  some normal
     constituents  of rabbit  semen during  repetitive  ejaculation.  Fertil. Steril.
     17:204-211,1966.

-------
                                   66

47.Paufler, S. K. and Foote, R. H.: Effect of triethylenemelamine (TEM) and--
     cadmium chloride  on spermatogenesis in rabbits. J. Reprod. Fertil. 19:
     309-319,1969.
48.Schwartz, D., Laplanche, A., Jouannet, P.  and David, G.: Within-subject
     variability  of  human semen in  regard to sperm count, volume, total
     number of spermatozoa, and length  of abstinence. J. Reprod. Fertil. 57:
     391-395,1979.
49. Katz, D. F. and  Overstreet, J. W.: Sperm motility assessment by videomicro-
     graphy. Fertil. Steril. 35(2): 188-193,1981.
50. Eliasson, R.:  Analyses of semen. In: Progress in Infertility, S. J. Behram and
     R. W. Kistnes, Eds., Little Brown and Co.: Boston; pp. 691-713, 1975.
51. Holtz, W. and Foote, R. H.:  Composition of rabbit semen and the origin of
     several constituents. Biol. Reprod. 18: 286-292, 1978.
52. Baker, M. W. G., Bremner, W. J., Burger, H. G., de Kretser, D. M., Dulmanis,
     A., Eddie, L., Hudson, B., Keogh, E.J., Lee, V.W.K.  and Rennie,G.:
     Testicular control of FSH secretion. Rec. Prog. Horm. Res. 32: 429, 1976.
53. de  Kretser, D. M., Kerr, J. B., Rick, K. A., Risbridger, G. and Dobas, M.:
     Hormonal  factor  involved in normal spermatogenesis. In: Testicular
     Development, Structure, and Function, A. Steinberger and E. Steinberger,
     Eds., Raven Press: New York; pp. 109-115,1980.
54.Psychoyos, A.:  Hormonal control  of ovoimplantation. Vitam. Horm. 31:
     201-256,  1973.
55.Bedford, J.  M.: Techniques and criteria used in the study of mammalian
     fertilization. In: Methods in Mammalian Embryology, J.C. David, Ed.,
     W. H. Freeman and Co.: San Francisco; pp. 37—63, 1971.
56. Yanagimachi, R., Yanagimachi, H. and  Rogers, B. J.: Use of zona-free animal
     ova as a  test system for assessment of fertilizing  capacity of human
      spermatozoa. Biol. Reprod. 15(4): 471-476, 1976.
57.Hahn,  J., Foote, R.  H.  and Cranch,  E. T.: Tonometer for measuring
      testicular consistency of bulls to  predict semen quality. J. Anim. Sci. 29:
      483-489,1969.
58. Oakberg, E. F.: Degeneration of  spermatogonia of the mouse  following
      exposure to X-rays, and stages in the mitotic cycle at which death occurs.
     J.Morph. 97:  39-54,1955.
59.Nicander, L.: Studies  on  the regional histology and cytochemistry of the
      ductus epididymis in rabbits.  Acta Morphol. Neerl. Scand. 1: 99—118,
      1957.
 60. Reid, B.  L. and  Cleland, K.  W.:  The  structure and  function of the
      epididymis. I. The  histology  of the  rat epididymis. Aust. J. Zool. 5:
      223-246,1957.
 61. Brooks, D.  E.:  Biochemical environment  of  sperm maturation. In: The
      Spermatozoon: Maturation, Motility, Surface Properties  and Comparative
      Aspects, D.W. Fawcett and J. M. Bedford, Eds., Urban and Schwarzen-
      berg: Baltimore; pp. 23-34,1979.
 62. Brandes, D., Ed.: Male Accessory  Sex Organs. Academic Press: New York;
      527 pp., 1974.

-------
                                   67

63.Hammerstedt, R. H.: Characterization of sperm  surfaces using physical
     techniques. In: Spermatozoon: Maturation, Motility, and Surface Proper-
     ties and  Comparative  Aspects, D.W. Fawcett and J.M. Bedford, Eds.,
     Urban and Schwarzenberg: Baltimore; pp. 205—216,1979.
64. Bedford, J.  M. and  Cooper, G.  W.: Membrane  fusion  events  in  the
     fertilization of vertebrate egg. In: Cell Surface Reviews, Vol. 5, Membrane
     Fusion, Elsevier North Holland: Amsterdam; pp. 66—111,1978.
65.Mann, T. and Lutwak-Mann, C.: Male Reproductive Function and Semen,
     Springer-Verlag: New York; 495 pp., 1981.
66. Hammerstedt, R. H.: Monitoring the metabolic rate of germ cells and sperm.
     In: Reproductive  Processes and  Contraception, K.W.  McKerns, Ed.,
     Plenum Publishing: New York; pp. 353-391, 1981.
67. Hegde,  U.  C., Shastry, P.  R. and Rao,  S. S.: Use of dithiothreitol for
     improved visibility of F-body in human Y-bearing spermatozoa. J. Reprod.
     Fertil. 53: 403-405, 1978.
68. Evenson, D. P.,  Darzynkiewicz,  Z. and  Melamed, M.  R.: Relation of
     mammalian sperm chromation heterogeneity to fertility. Science 210:
     1131-1133,1980.
69.Gledhill, B. L., Lake, S. and  Dean, P. N.: Flow cytometry and sorting of
     sperm and  other male germ cells. In:  Flow  Cytometry and Sorting,
     M.Melamed, P.Mullaney, and M.Mendelsohn, Eds., J. Wiley and Sons:
     New York; pp. 471-484,1979.
70. Rudak,  E.,  Jacobs,  P. A.  and   Yanagimachi,  R.:  Direct  analysis of
     chromosome constitution of human spermatozoa. Nature 274: 911—913,
     1978.
71. Aafjes,  J. H., Vels, J. M. and Schenck, E.:  Fertility of rats with artificial
     oligozoospermia. J. Reprod.  Fertil. 58(2): 345-352, 1980.
72.Desjardins, C., Kirton, K. T.  and Hafs, H. D.: Sperm output of rabbits at
     various ejaculation frequencies and their use  in the design of experiments.
     J. Reprod. Fertil. 15: 27-32, 1968.
73. Seidel, G. E., Jr. and Foote, R. H.: Variance components of semen criteria
     from  bulls  ejaculated  frequently and  their use in experimental design. J.
     Dairy Sci. 56: 399-405,1973.
74. MacLeod, J.: Human male infertility. Obstet. Gynecol. Surv. 26: 335—351,
     1971.
75. Janick, J. and MacLeod, J.:  Measurement of human spermatozoan mobility.
     Fertil. Steril. 21:  140-146,1970.
76. Lubs, H. A.:  Testicular size in Kleinfelter's Syndrome in men over fifty.
     Report of a case with XXY/XY mosaicism. New  Engl. J.  Med. 267:
     326-331,1962.
77. Asbjornsen, G., Molne, K., Klepp, O. and Aakvaag, A.: Testicular function
     after  combination chemotherapy for Hodgkin's disease.  Scand. J. Hae-
     matol. 16(1): 66-69,1976.
78..Van Thiel, D. H., Sherins, R. J.,  Meyers, G. H. and Devita^ V.T.,  Jr.:
     Evidence   for a  specific seminiferous tubular factor  affecting follicle-
     stimulating hormone secretion in man. J. Clin. Invest.  51: 1009—1019,
     1972.

-------
                                   68

79. Cunningham, G. R. and Huckins, C.: Serum FSH, LH, and testosterone in
      60Co 7-irradiated male rats. Radiat. Res. 76: 331-338,1978.
80. Verjans, H. L. and Eik-Nes, K. B.: Hypothalamic-pituitary-testicular system
      following testicular  X-irradiation. Acta  Endorcinol. 83(1):  190-200,
      1976.
81. MacLeod,  J.:  Human  seminal cytology  following the administration of
      certain  antispermatogenic  compounds. In: Agents  Affecting Fertility,
      C. R. Austin and J. S. Perry, Eds., Little Brown and Co.: Boston; pp.
      93-123,1965.
82. Belsey, M. A., Eliasson, R., Gallegos, A. J., Maghissi, K. S., Paulsen, C. A.
      and Prasad, M. R.N.: Laboratory Manual for the Examination of Human
      Semen and Semen-Cervical  Mucus Interaction, Press Concern: Singapore;
      43 pp., 1980.
83. National Academy of Sciences: Drinking Water and Health, Vol. 3, National
      Academy Press: Washington, D.C.; pp. 25-265,1980.
84. Young, W. C., Ed.: Sex and Internal Secretions, Vol. 1,3rd Edition, Williams
      andWilkins: Baltimore;pp. 161-448,1961.
85. Young, W. C., Ed.: Sex andlnternal Secretions,Vol. 2,3rd Edition, Williams
      andWilkins: Baltimore;pp.707-796,1173-1239,1961.
86. Mann, T.: The Biochemistry of Semen and of the Male Reproductive Tract,
      Methuen: London; 240 pp., 1964.
87. Hamilton, D. W. and Creep, R. O., Eds.: Handbook of Physiology, Section 7,
      Vol.  5:  Male  Reproductive System, American  Physiological  Society:
      Washington,D.C.; 519 pp., 1975.
88. Creep, R.  0., Koblinsky, M.  A. and Jaffee, F. S., Eds.: Reproduction and
      Human Welfare: A Challenge to Research, MIT Press: Cambridge, Mass.;
      622 pp., 1976.
89.Wyrobek, A. J. and Gledhill, B. L.:  Human semen assays for workplace
      monitoring. In: Proceedings' of Workshop on Methodology for Assessing
      Reproductive Hazards  in  the Workplace. Center for Disease  Control,
      National Institute for Occupational Health  and Safety; pp. 327—355,
      1980.
90. Gledhill, B. L. Personal communication, Lawrence Livermore Laboratory:
      California; 1981.
91. Beatty, R. A., Bennett, G. H., Hall, J. G., Hancock, J. L. and Stewart, D. L.:
      An  experiment  with heterospermic  insemination in cattle. J. Reprod.
      Fertil. 19: 491-502,1969.
92. Overstreet, J. W. and Adams,  C. E.:  Mechanisms of selective fertilization in
      the rabbit: sperm transport and viability. J. Reprod. Fertil. 26: 219—231,
      1971.
93. O'Connor, M. T., Amann, R. P. and Saacke, R. G.: Comparisons of computer
      evaluations of spermatozoa! motility with standard laboratory tests and
      their use for predicting fertility. J. Animal Sci.; in press, 1981.

-------
                        APPENDIX
         DETAILS OF TEST PROTOCOLS AND
          GLOSSARY OF TERMS FOR MALE
                   RISK ASSESSMENT


   I. DESCRIPTION AND DISCUSSION OF TESTS USEFUL
               IN ANIMAL MODELS OR MAN

                        Body Weight

   Measure body weight  of all test animals weekly starting two
weeks before  administration of the compound and continuing until
termination of the study.
                   Testicular Characteristics
Testis size in situ
   The number of spermatozoa, and to a lesser extent the quantity
of testosterone, produced by the testes of normal individuals is a
function of testis size and, to a lesser extent, of variation in the
proportion  of  the  testis  composed  of germinal  elements  and
interstitial tissue (1). Therefore, assessment of testicular size is very
important from a functional standpoint. In scrota! animals, testicular
size can be  measured easily,  accurately, repeatedly and without
damage to the individual (2—5). In many species of laboratory and
domestic  animals, testis  size is correlated (correlation  coefficient
r = 0.8—0.9) with sperm output in ejaculated semen when males are
ejaculated frequently (e.g., four ejaculates per week) (1, 2). Changes
in  testis size  should  be  correlated with results  of other tests to
increase the accuracy of the analysis.
   Measurements of testis size should be made biweekly or weekly
with animal models and could be made part of an annual physical
examination given to men working in a hazardous environment.
                             69

-------
                               70

    Scrotal circumference. This measurement can be taken easily in
animals with  pendulous  scrotum  (4).  To  reduce variation, the
measurements should be made with a standard procedure.
    Linear  measurements. Length and  width can be  measured in
species such as dogs, rabbits, bulls, and horses (1, 2—5). Length and
width measurements  are correlated  with testis weight O0.90). If
these data are correlated with seminal characteristics, adjustment for
time  lag  in  spermatogenesis  and  sperm  transport  through the
excurrent ducts is necessary.

Testis weight
    Each testis must  be dissected  free from the epididymis and
pampiniform plexus, and weighed when model animals are killed or
castrated. Testis weight, relative  to  norms for that breed or strain,
can reveal gross differences resulting from a treatment.

Spermatid reserves
    Counting of homogenization-resistant spermatid nuclei in testicu-
lar homogenates is a  simple, accurate, and sensitive method for
measuring sperm production. This method  can be accomplished with
simple equipment and does  not require extensive training. The nuclei
of elongated  spermatids are resistant to  mechanical  and chemical
disruption and are easily  identified  after physical  disruption  of
testicular tissue (1,6, 7). With human  testes, small biopsies can be
used (8), and the tissue should be fixed in  glutaraldehyde before
homogenization, because some spermatid nuclei may not be fully
condensed  (9).  The  interval  from  when spermatids acquire the
resistance  to homogenization  until  spermiation is  a constant for a
 species or strain (1). Thus, the number of resistant spermatids is a
 direct measure of  the production of spermatozoa by  the testis and
 the survival of the precursor spermatogenic cells (1).
    Either biopsy material (20 mg or more), a representative sample
taken at necropsy, or the entire testes (for rats and rabbits) can be
homogenized or disrupted ultrasonically (6, 10). Resistant spermatid
nuclei are  counted in a cytometer (at least 6  chambers per sample).
 Counts  should be expressed on   both  a  per-testis  basis and  a
 per-mg-of-parenchyma  basis (11).  Counts from  treated  animals
 should be compared to those for concurrent control males.
    The time of this analysis relative to an acute  treatment or the
 onset of chronic  treatment can be varied so as to reveal  possible
 damage to cells in specific stages  of spermatogenesis. The interval

-------
                               71

between onset of treatment and evaluation should be expressed in
terms of the duration of one "cycle of the seminiferous epithelium"
for that species. Preferred times for evaluating agents should  be
chosen according to the kinetics of spermatogenesis (7, 12—17).

Histopathological analysis of testes

    Histologic  analyses of testicular biopsies or whole testes must be
performed  on  animal models  and, in special  cases,  could  be
performed  on man.  Qualitative and quantitative analyses  of in-
creasing  complexity  yield  general  or  precise information. It is
axiomatic that serious disturbance will  be evident to the observer
using direct simple evaluations of germinal epithelium in histological
preparations of whole testes. Threshold effects require a detailed
evaluation such as that recommended below. Electron microscopy is
not  considered to  be useful  for screening of damaging  agents.
Testicular tissue must be fixed  immediately in Bouin's or Zenker's
fluid (10% formalin is not satisfactory). Slides should be stained with
hematoxylin and eosin for simple analyses and with periodic-acid-
Schiff-hematoxylin if a more precise  determination of the stages of
spermatids is required (17).
    Gross morphology. Appearance of Leydig interstitial cells (18);
occurrence of  lymphoid  cell  or  macrophage infiltration (19);
presence of germ cells of each stage (spermatogonia, spermatocytes,
spermatids, sperm) in seminiferous tubules (20); presence of large
numbers of degenerating (21), multinucleate (22), or abnormal germ
cells (23) should be noted.
    Nonfunctional tubules. The  percentage of tubular cross-sections
with no  evidence of spermatogenesis  (i.e., <4 germ cells)  should be
scored during  brief examination at 100X or 400X magnification of
250 cross-sections per testis (24, 25). Such examination  could  be
performed 2 to 7 days after acute treatment or 6 cycles after onset
of chronic  treatment.  The integrity of the layer of Sertoli  cells in
these sterile tubules should also be noted.
    Tubules with spermatids  lining the lumen. The  end product of
spermatogenesis is reflected in the "mature" spermatids about to be
released from the Sertoli cells. Tubules with spermatids aligned at the
lumen can be  easily recognized (16). The incidence of such tubules is
a  characteristic  of  the  species.  Deviations between control and
treated males reflect testicular dysfunction.
    Seminiferous tubule diameter. Diameters can change with inter-
ference of  tubular function (26).  Measurements of minor diameter
should be  taken on essentially  round tubule cross-sections  (cut at

-------
                               72

right angles to their long axis) taken from several different locations
in sections used for scanning other aspects of gross histology. Only
tubules in which the minor diameter is within  10% of the major
diameter  (i.e.,  the sections  are  essentially  transverse)  should be
measured.

Counts of preleptotene or leptotene spermatocytes
    The number of leptotene spermatocytes  per  Sertoli  cell with a
visible nucleolus can  be quantitated because of a characteristic
nuclear morphology of leptotene   spermatocytes  (27, 28).  The
number of leptotene spermatocytes and the number of Sertoli cells
with a visible nucleolus should be determined in the same set of
tubules. The ratio  of  spermatocytes per Sertoli  cell is  a  sensitive
measure of testicular damage; effects  of as little as 5 rad of radiation
can be detected (20).

                    Epididymal Characteristics

Weight of distal half of epididymis
    The distal portion of the epididymis can  be isolated by severing
the corpus epididymidis midway between the  caput and cauda and at
the junction of the distal cauda with  the ductus deferens. The distal
epididymis  and  the contralateral epididymis should be  weighed
promptly.

Number of sperm in the distal half of epididymis
    One epididymis, weighed as above, is homogenized to liberate the
spermatozoa contained therein (6, 7); simple  mincing of the tissue is
inadequate. Sperm cells are counted using a cytometer  (at least 6
chambers counted per sample). The  results should be expressed as
total counts. The epididymis evaluated could be alternated within
each  control or treated  group to ensure representative sampling if
there is any systematic difference between sides.

Motility of sperm from the distal end
    Sperm from the distal end of the remaining cauda epididymidis
will be expressed into a phosphate-buffered saline solution contain-
ing 5 mM of glucose or pyruvate plus 0.1%  bovine  serum  albumin,
polyvinyl alcohol, or similar macromolecules. Sperm concentration
should be standardized  to  10 X  106 to 40 X 106 per ml,  and the

-------
                                73

 percentage  of motile  sperm determined at  37°C under conditions
 similar to  those described for  estimating  percentage  of motile
 spermatozoa in ejaculated semen.

 Gross morphology of spermatozoa from the distal end

    The same semen preparation used for estimating the percentage
 of motile spermatozoa will be viewed by phase-contrast microscopy
 at 400X for evaluation of gross morphology. The proportion of the
 spermatozoa from treated animals, in comparison with controls, with
 misshapen heads, acrosomal defects or distorted swimming patterns,
 will be estimated.

 Detailed morphology of spermatozoa from the distal end

    For detailed evaluation of  sperm morphology, smears will be
 prepared by a procedure minimizing artifacts (29), then stained with
 eosin-nigrosin (or other differential  stain). A total of 200 to 400
 spermatozoa should be classified per sample. Smears can be preserved
 as  a  permanent record,  pr videotapes can be  prepared. Detailed
 morphological or morphometric examination is possible with either.

               Accessory Sex Gland Characteristics

    1. The accessory sex glands are biomonitors of androgen produc-
 tion by the  testes. Thus, accessory  sex gland weight will be recorded
 when each male animal is killed.
    2. For rats, the vesicular glands are discrete organs and very easily
 distinguishable. After removal and expression of the viscid fluid, the
 glands should be blotted and weighed.
    3. The individual accessory  sex glands  are not discrete in the
 rabbit (30).  Thus, the  total set of accessory glands will be excised as
 a single unit, blotted, and weighed.  The organs will be reweighed
 after removal of any secretion present in the vesicular glands.

                        Seminal Analysis

 General aspects of seminal analysis

    (a) Analysis of semen offers  a convenient approach for monitor-
ing function of the germinal epithelium and, with less specificity, the
functions  of the epididymides,   prostate,  vesicular glands,  and
bulbourethral glands (11). An abnormality in  epididymal function
may be detected in semen ejaculated  3 to 15 days after epididymal

-------
                                74

dysfunction. An abnormality in spermatogenesis typically cannot be
detected  in  semen until after  at least  1  to 4  cycles  of  the
seminiferous  epithelium (16-64  days in  man or 11-43 days in
rabbit) have passed (1, 11, 14,31), plus time for epididymal trans-
port. This long interval is required because an agent must accumulate
to a toxic concentration and produce a  lesion in germ cells at  a
specific  point in  their  development (often  >2  cycles  of  the
seminiferous  epithelium before the end of spermatogenesis) before
production of more mature germ cells is affected. After passage of a
given interval,  evidence of the lesion can  be seen within the testis,
but the lesion will not be evident in semen until the affected germ
cells have completed spermatogenesis  (2—3 cycles), passed through
the  epididymis (4—16  days), and  appeared  as  spermatozoa in
ejaculated semen.
    (b) Multiple samples  can  and  should  be  obtained from  each
individual male. Both quantitative and qualitative characteristics of
more than one ejaculate must be evaluated to gain a reasonable
understanding  of  testicular function (1, 11,32). Data for  samples
collected before experimental exposure can be used as one basis for
assigning males to control or  treatment groups or as a covariant in
the statistical analysis.
    (c) The species, strain, and age of males; testicular size; season;
method  of  semen  collection;  and  interval  since  the  previous
ejaculation(s) all influence quantitative characteristics of semen and
must be carefully controlled (11,  33).
    (d) If seminal  analyses are planned, use of a species from which
semen can be collected by artificial vagina or  digital manipulation
(masturbation) is  essential. Suitable species include man, rabbit, dog,
bull, and minipig (1,2, 11,32-34).  The rabbit is the species of
choice   for  screening potentially  toxic  agents because  of size,
availability, cost, and ease of use. Small rabbits (e.g., Dutch Belted)
are  as  good  as  larger breeds (e.g., New  Zealand White)  and are
cheaper to house. Rams, goats,  and stallions are less ideal because
seasonal changes are more profound. Subhuman primates probably
will not be  used  frequently  because  of their limited numbers and
cost. Although useful in many other aspects, rats have limited use (as
do  mice) because  of their  small  seminal volume,  difficulty in
quantifying seminal characteristics, and the necessity to use  electro-
stimulation for semen collection. Improved procedures for quantita-
tive collection of  semen from rats or mice are unlikely to overcome
their limitations.  However,  as  outlined  above,  cauda epididymal
sperm can be obtained (on a one-time basis) from a mouse or rat and

-------
                                75

evaluation of the motility and morphology of epididymal sperm is
desirable.
    (e) For most studies, sexually mature males (body weight <90%
of maximum value for that strain) should be used. If consequences of
exposure before puberty are to be evaluated, the age or body weight
when a given number of sperm are first ejaculated may be a useful
criterion  (requires  at least weekly testing) and  the  postpubertal
changes in semen quality could be monitored (33).
    By  monitoring seminal characteristics longitudinally from  ex-
posure,  through a reasonable interval when an  effect  might be
expressed  (6 X the  duration  of the  cycle  of the  seminiferous
epithelium for the  species studied) and during a recovery phase (if
appropriate)  of twice this duration, information  can be obtained on
the  point when damage is expressed and when  recovery occurs
(1, 2, 14).

Volume

    (a) Volume of the ejaculate should be measured with an accuracy
of greater than 90 to 95%. To measure accurately ejaculates with a
small volume, it is recommended that collection tubes be preweighed
and  ejaculate volume be calculated from the weight,  assuming a
specific gravity of 1.0.
    (b) Systematic errors associated with seminal loss during collec-
tion  or   transfer  to a  measuring device  should  be  minimized.
Measurement  of ejaculate volume within the collection vessel, after
addition  of  a known volume of buffer if essential,  is  desirable.
Systematic  errors  in  measurement often  can be corrected for
(1,2,11).
    (c) If a uniform collection interval and standardized collection
procedure are used (1, 11), differences  of >25% in ejaculate volume
probably  could be detected in a longitudinal study utilizing ten
rabbits per treatment group (35).  A  difference of this magnitude
probably  would reflect  abnormal  function of  the accessory sex
glands if unaccompanied by a change hi  sperm output.
   (d) The coefficient of variation for  volume of a human ejaculate
is unknown but could be calculated from  available data. It is likely
that a sizable number of ejaculates must be evaluated  for  each
individual in a group to detect a 25% change in seminal volume.

Seminal plasma constituents

   (a) The biochemical components of seminal plasma may reflect
the functionality of the epididymides and accessory sex glands (11),

-------
                               76

but the concentration of a compound in a seminal plasma has limited
diagnostic value (11).
    (b) If collection procedures are rigidly standardized for a species,
a  marked  change  (>25%) in  the  total  mass  of a  constituent
ejaculated,-could reflect the function of the excurrent duct system or
one or more accessory sex glands. It is not clear at present, however,
whether  any constituents change their concentration markedly in the
course of repeated ejaculations with humans (see Research Needed).

Spermatozoal concentration
    (a) The term spermatozoal concentration is preferred to those of
sperm count or sperm density.
    (b) Sperm concentration,  by itself, provides little  information
(11), but sperm concentration must be determined accurately so that
the total number  of sperm  per  ejaculate can be  calculated (see
below).
    (c) Sperm concentration should be determined using a calibrated
spectrophotometer or electronic cell counter,  if contaminating cells
or debris are not a problem, because of their accuracy and precision
(2, 11). If extraneous material is present in the semen, visual counts
using  a cytometer are  essential. Use  of a cytometer  (with  a
phase-contrast microscope) is time  consuming, and  >6  replicate
counts are necessary to achieve >90% accuracy for a single sample.

Total sperm per ejaculate
    (a) The   term  total  number  of  sperm  per  ejaculate  (vol-
ume X sperm  concentration)  is preferable to that  of total sperm
count.
    (b) Total  sperm per ejaculate represents  the number of sperm
coming,  from the  excurrent duct system and  is independent of the
degree of dilution by accessory sex gland  fluid (11).
    (c) When semen is  collected by  a uniform procedure and  total
sperm per  ejaculate  is  averaged over  time,  daily  sperm output
(number of sperm in a series of ejaculates divided by the time span)
can be calculated. Daily sperm output, in rabbits and bulls, is highly
correlated (sO.9) with daily sperm production (1, 11, 36).
    (d) To measure daily  sperm output accurately (1, 11), a uniform
interval  of one,  two,  or  three days  between semen collections  is
essential,  and  the  series of  ejaculates  should extend  over  14
(preferably 20) days (data for the  first 3—6 ejaculates should  be
 excluded a priori and data for the remaining >6 samples averaged).

-------
                                77

    (e) If semen is collected infrequently (one ejaculate weekly), a
50% reduction in  sperm production probably would be undetectable
(11). To  have a 75% chance of detecting a difference of 50% daily
sperm output would require about 20  rabbits per treatment and
ejaculation for >5 weeks (35).
    (f) A  formula  relating the coefficient of variation (CV) between
counts; the significance  level at which the statistical test is to  be
performed,  a (e.g., a = 0.05,  a =0.01);  the desired power  or
sensitivity of  the statistical test, 1-/3 (e.g., 1-/3 = 0.50, l-j3 = 0.90); and
the  sample size  of  the study, N (i.e., N treated  and N control
animals),  to the required change (in terms of percent of 'normal' or
control values) in the test criteria is given by
                                   = % change
                       VN/2
where Za = 1.645 for a = 0.05 and Za = 2.326 for a = 0.01, and
Zj.0 = 0, 0.253, 0.524, 0.842, and 1.281 for 1-0 = 0.5, 0.6, 0.7, 0.8,
and 0.9, respectively. It should be noted that this formula assumes a
one-sided statistical test, that is, looking for changes between treated
and control animals in only  one direction (e.g.,  decrease in sperm
concentration).  Along with  the coefficients of  variation given  in
Table 10, it can be used to determine the adequacy of different
experimental designs. For example, the largest coefficient of varia-
tion,  other than  for  accessory sex gland weight, is for the test
criterion total sperm/ejaculate in Dutch Belted rabbits, CV = 0.75.
Assuming that any statistical comparison between 12 treated and 12
control rabbits is 'conducted at the a = 0.05 level (i.e., 5% test level),
to have at least a 50% chance of detecting a statistically significant
difference (i.e., power = 0.50), then the treatment must produce  at
least    a    50%    change   in  the   test  criterion,  that  is,
(1.654 + 0)(0.75) V 12/2 = 0.5.  Because each of the  other criteria,
except for accessory sex glands, have coefficients of variation of less
than 0.75,  they would have  the same power, 50%, of detecting a
smaller  effect; for example, for testis weight, CV = 0.2 giving a
percent change of 13%, (1.645 + 0) (0.20)A/12/2 = 0.13.

Sperm motility

    (a) Rigid control of temperature at 37°C and other conditions of
evaluation are essential (2, 11).
    (b) Visual evaluation of sperm motility  using diluted semen and a
phase-contrast microscope is informative  and rapid,  although sub-

-------
                               78

jective. Visual estimations are adequate for an initial screen, provided
that control and treatment samples are presented randomly in a blind
manner to the  observer. For second- or third-level analysis, more
objective  procedures, such  as  videotape  and analyses or  track
motility (37—39) should be considered.
    (c) The percentage of progressively motile sperm, the translatory
velocity, and the presence of sperm moving in a circular pattern or
backward should be recorded.
    (d) A reduced percentage of motile sperm might reflect abnormal
spermatogenesis, abnormal functions of the epididymis or entrance
of  an antimotility  factor into the semen (via  the  excurrent ducts,
prostate,  bulbourethral  glands,  or vesicular glands) where it could
exert a direct effect on the sperm.
    (e)The  percentage  of  motile   sperm  probably  reflects both
normality of spermatogenesis and sperm metabolism.
    (f) Variation within  males in the percentage of motile  sperma-
tozoa (and probably velocity) is less than  for ejaculate volume or
total sperm per ejaculate (35, 40).
    (g) A significant  decrease in  the percentage of motile sperm
would be a strong indicator for a potential decline in fertility  and
especially so if sperm numbers are limited.

Spermatozoal morphology
    (a) Abnormalities of sperm  morphology reflect dysfunction of
the germinal epithelium (primary abnormality) or of the excurrent
duct system (secondary abnormality). Certain  abnormalities cannot
be clearly attributed to a specific site of action.
    (b) An  increase  in  the percentage  of abnormal sperm  may
precede a decline in the total number of sperm per ejaculate (if any)
and can  serve as a sensitive indicator  of  epididymal or testicular
function.
    (c) Within a male,  sperm morphology  is quite consistent over
time (41). This consistency makes sperm  morphology a sensitive
probe while requiring fewer samples per male for an experiment of a
given precision.
    (d) Evaluation of sperm morphology is subjective (42) and must
be carefully standardized among laboratories (11, 42, 43). A detailed
classification probably is unnecessary in an  initial screening process.
Classification of spermatozoa, based on light microscopy, as normal
or abnormal head,  normal or abnormal tail  is adequate for a screen.
For second- or  third-level screening, a more complex classification
might be  used (42-44).

-------
                               79

    (e) Evaluation of sperm morphology using wet preparations and
phase-contrast  microscopy is recommended  for  simplicity  and
freedom from artifacts (11, 29), although preparation and retention
of stained smears (a simple stain like eosion-nigrosin or eosin-analine
blue) is desirable for archival purposes.

Ejaculated sperm as an in vitro test system

    (a) Substances can pass from blood into semen via the fluid from
the.  excurrent  ducts and  accessory   sex  glands  and could  be
spermicidal or alter sperm function.
    (b) Agents can be screened economically by incubating sperm in
vitro under standard conditions  in  a  protein-containing  buffer at
37°C for 4 to 8 hours. Sperm could be exposed to the agent briefly
(10—30 minutes)   or throughout the  incubation period.  A  dose-
response curve should  be established using objective methods of
evaluation  and sperm from humans or other species (rats, rabbits,
dogs, or bulls).
    (c) The decline in percentage of motile sperm over time is an
exceUent criterion, but other criteria (e.g., integrity of the acrosome
and  plasma  membrane,  oxygen  consumption,  adenosine  5'-
triphosphate content, or degree of agglutination [45]) could be used.
    (d) Compounds that are spermicidal in  vitro at concentrations
that could be anticipated or shown to be present in blood or seminal
plasma should be carefully screened in vivo. Failure to demonstrate a
spermicidal action in vitro is  not evidence that an agent would be
free of effects on male reproduction, nor is spermicidal  action in
vitro evidence of in vivo  activity.
            Assessment of Male Reproductive Toxicity
                 Using Endocrinological Methods

General

    (a) This  section describes general aspects of applying endocrino-
logical  methods  to  the study  of  male  reproduction.  Specific
applications of these  techniques to studies in animals and men are
described elsewhere in this account.
    Normal  male  reproductive function requires hormonal stimula-
tion of the testes and production by the testes of adequate numbers
of sperm and the hormone testosterone. Luteinizing hormone (LH)
and follicle-stimulating hormone  (FSH)  are  the  two  hormones
necessary to maintain normal testicular function.  These hormones

-------
                               80

originate in the pituitary gland and travel through the blood to affect
the testis. The production of LH and FSH and their release from the
pituitary are stimulated by gonadotropin-releasing hormone (GnRH),
which  is produced in the hypothalamus  at the base of the brain.
Testosterone and other hormones produced by the testis are carried
by the blood throughout the body. At the pituitary, these hormones
tend to decrease the production of LH and FSH,  that is, they exert a
"negative feedback" effect on LH and FSH secretion.
    (b) If a defect occurs in hypothalamic  or  pituitary function,
blood levels of FSH and LH will tend to decrease.
    (c) If a defect occurs in the testis (either in sperm or testosterone
production), FSH and LH  levels will tend to increase because of lack
of the "negative feedback" effect of testicular hormones.
    (d) In addition to its effects on the pituitary, testosterone exerts
many  effects throughout the body. It is necessary for expression of
male  sexual  behavior  and the ability  to perform  intercourse,
stimulates muscle  and bone  development  and  red  blood cell
production, and is essential for many other aspects of normal body
function. A decrease in blood levels of testosterone can be expected
to affect all these functions adversely.
    (e) It is clearly established in all mammalian species investigated
that an endocrine defect in the brain, pituitary,  or testis may inhibit
spermatogenesis and normal sexual behavior and cause sterility. Less
severe defects in these tissues (not so severe as to lead to infertility)
might be detected by measurements of hormone concentration in the
blood. In certain situations,  including studies of human beings,
hormone measurements  are very  practical, because  they can be
performed on ordinary samples of blood serum, whereas seminal
 fluid or testicular tissue may be difficult or impossible to obtain.
    (f) Hormonal  measurements are important and sensitive tools in
 the assessment of toxicity to the male reproductive system. They can
 be compared directly among a variety of species and between control
 and treated groups of any species including man. Hormonal data may
 give a clue as to the tissue in which a toxic effect is occurring.

 Hormone assay and application
    (a) Hormones  commonly  are measured  by  radioimmunoassay.
 This  is  an  extremely  sensitive technique  and, when done  in  a
 competent laboratory, is  quite reliable for measuring  testosterone,
 FSH,  and  LH.   Many  commercial laboratories  perform  radio-
 immunoassays  for LH, FSH, and  testosterone in human  blood
 samples.

-------
                                81

    Developing these assays requires familiarity with the technique,
 access  to  counters  for  radioactivity  and availability of specialized
 assay reagents  such as  purified preparations of the hormones and
 antibodies to these hormones.
    (b) Testosterone is an identical molecule in all species, so it can
 be measured  from  any  species in  a  single  assay.  Nevertheless,
 appropriate  species  controls are essential to preclude the possibility
 that a  cross-reacting molecule is altering test results. Both LH and
 FSH are protein hormones that differ slightly in molecular structure
 among  species.  Assays  for LH and  for FSH  tend to be species-
 specific, so that assay reagents appropriate for the species in question
 generally are required for measuring LH or FSH. For rats and man
 these reagents  are widely available, and such reagents have recently
 been introduced for rabbits. For normal adult male rats, however,
 the  concentration of LH in peripheral blood is usually below the
 sensitivity level of available radioimmunoassays.
    Measurement of  blood  levels of hormones  yields  a direct
 assessment of  the level of  exposure  necessary to produce a toxic
 effect. The methodology is sufficiently accurate to detect changes of
 20% in mean  hormone  levels using generally available numbers  of
 animals (e.g.,  20 per group). Statistical adequacy  for these assays
when performed competently is very  good; hi general, within-assay
coefficients of variations are below 10%, and between-assay coeffi-
cients of variations are less than  15% (46). These assays are specific
measures of reproductive toxicity; disease of other organ systems will
not  affect these  measurements unless the disease concomitantly
affects the reproductive system.
   Any statistically  significant difference between hormone levels  in
a comparison of control and exposed animals or men can be accepted
as strong evidence for a toxic effect of the exposure on reproduction.
If such  an effect were established by animal studies, this could be
used as strong  evidence  that a similar effect would occur in human
beings. In essentially every instance studied in detail, agents found to
be testicular toxins  in one  species  have a  similar  effect in other
species (47).
   (c) Hormone levels may be measured in single blood samples,
although evaluation of several samples taken at two-hour intervals is
better. The time of  day should be standardized because of diurnal
rhythms. Ordinarily, no  special preparation  is necessary concerning
diet or physical activity.
   (d) For all known reproductive toxins, the damage to reproduc-
tion is reversible if the exposure level is so low as to produce only a

-------
                               82

minimally detectable effect. With a more severe insult or a prolonged
toxin exposure, the damage may be irreversible.

             Examination of Known Toxic Exposures

Humans
    Seminal fluid analyses. At least one and preferably five seminal
fluids  obtained by  masturbation  at  two-day intervals should be
submitted  by each man.  Precautions must be taken to ensure an
accurate measurement  of  seminal volume  (11). The age  of  the
individuals and the abstinence interval between samples should also
be  considered in the evaluation (48-50). Variability  among ejacu-
lates from the  same  individual  is influenced  by  the length  of
abstinence (50). The semen should be analyzed for volume, sperm
concentration,  and  total  sperm  per ejaculate.  Total sperm  per
ejaculate  must  be calculated and compared with norms. Sperm
motility (at 37°C) should be objectively assessed using phase-contrast
microscopy; motility should be characterized in terms of percentage
of  motility and velocity.  If feasible, videotapes should be made of
the living sperm cells, with the tapes being subsequently analyzed in
a laboratory familiar with this technique. Seminal smears should be
fixed (29) for subsequent analysis. Tests of sperm function such as
penetration of zona-free hamster  eggs may not be feasible for field
work. However, if persistent infertility remains undiagnosed after
completion of the other studies proposed, the hamster egg in vitro
penetration test (51) should be considered.
     Blood hormone levels. Peripheral venous blood samples should be
obtained  at a  standardized time of day (preferably  0700-0900
hours) for measurement of serum  luteinizing hormone (LH), follicle-
stimulating hormone (FSH), and testosterone by radioimmunoassay.
 Hormone  measurements on both the exposed  and control groups
 should be performed in the same laboratory. This laboratory must be
 one that  is recognized for reliability and  that maintains careful
 quality control records.
     Gonadotropin-releasing  hormone  (GnRH)  test.  This test has
been  demonstrated  to be  capable of  detecting mild degrees of
primary testicular dysfunction insufficient to elevate basal hormone
levels out of the normal range (52). It is not necessary if the basal,
 unstimulated-hormone  levels are abnormal. Blood samples for mea-
 surement  of LH and FSH are obtained before and at 30, 60, and 90
 minutes after administration of GnRH (100 Mg i.v:). Synthetic GnRH
 is now available for use as an investigational new drug through several

-------
                                83

 pharmaceutical  companies and will probably  be approved by the
 Food and Drug Administration for general use.

 Animal models

    Rats. Twenty mature male rats of a highly fertile strain (body
 weight must be >90% of adult normal for that strain) will be given
 >0.5 of the maximum tolerated dose (MTD) by inhalation,  intra-
 peritoneal (i.p.) injection, drinking water, or gavage for a period of
 exactly  6  cycles  of seminiferous  epithelium  (12.9  days  per
 cycle X 6 = 77 days). An  appropriate control group (or groups) will
 be evaluated concurrently.  If the  agent is given by injection  or
 gavage, both nonhandled and vehicle-injected control groups are
 necessary. Each rat will be weighed weekly  starting 14 days prior to
 initial dosing (day —14) and continuing to day 78. Each male will be
 caged with two  sexually mature, virgin female rats between days 65
 and 71  (for a total of 6 nights) to evaluate fertility. On day 78, blood
 will be taken by cardiac puncture immediately  (<1 minute)  after
 removing a male rat from his cage in the animal room and the male
 rat killed. Serum will be frozen.
    Both testes will be weighed. One will be fixed in Bourn's fluid for
 histologic examination and determination of  the  number of lepto-
 tene spermatocytes per Sertoli cell nucleolus. The second testis will
 be homogenized and the number  of resistant  spermatid nuclei
 determined. The vesicular  glands  will  be  weighed as an  indirect
 measure of circulating testosterone concentration. The distal half of
 one epididymis (half corpus plus cauda) will be weighed. Sperma-
 tozoa  will  be expressed  from the severed  end of the distal cauda
 epididymidis into phosphate-buffered saline containing 0.1% bovine
 serum albumin.  The percentage of progressively motile spermatozoa
 will be determined  under phase-contrast microscopy and the inci-
 dence of abnormal  spermatozoa recorded. A stained slide of the
 spermatozoa will be made for documentation. The distal half of the
 contralateral epididymis will be isolated, homogenized, and the total
 number of sperm heads determined. Concentration of testosterone,
 LH, and FSH in serum will be determined.
    The females will  be  killed  on day 83 to 89  (18  days  after
 mating), and the numbers of corpora lutea and implantation sites, as
 well as embryo viability, will be determined.
    Rabbits. Twelve  mature male rabbits  (body weight  must  be
 >90% of adult normal for that strain) will be given the >0.5 MTD by
.inhalation,  i.p.  injection, or in drinking water (the same route  of
 administration  should be used for both  rats and rabbits  when

-------
                                84

possible) for a period of 6 cycles of seminiferous epithelium (10.7
days per cycle  X 6 = 64 days).  An appropriate control group (or
groups) will be established concurrently. Testis size and body weight
will be measured weekly starting on day -14 and continuing through
day 65. Two ejaculates will be collected every 3 to 4  days (e.g.,
Monday and Thursday) using an artificial vagina. The volume, with
and without gel, concentration, and total sperm per ejaculate will be
measured.  Sperm  motility  and gross sperm  morphology will be
determined using  phase-contrast microscopy. Libido  will be  sub-
jectively assessed weekly. Each male will be mated with two virgins,
sexually mature females over a 4-day period (between days 54 and
57) and the females allowed to kindle. On day 65, blood will be
taken by cardiac puncture and  the male rabbits killed.  Evaluation
will be similar  to  that for rats. Testis size and weight,  number of
homogenization resistant spermatids,  weight  and sperm  content of
the distal epididymis, and motility and morphology of sperm from
the distal epididymis will be evaluated. Gross histologic evaluation
and enumeration  of the number of leptotene spermatocytes per
Sertoli cell nucleolus will be made on one  testis fixed in Bouin's
fluid. Weight of  the accessory  sex glands will be  recorded as an
indirect measure of circulating-testosterone level. Blood will be saved
for possible assay for FSH, LH, and testosterone.
    Pregnancy rate and litter size will be determined for females bred
 to each male.

                          Fertility Testing

 Tests available
     Humans. In vitro  oocyte penetration  tests are  the  only means
 available for assessing the fertilizing capacity of human sperm. Since
 human in vitro fertilization cannot itself be used as a test, substitutes
 must be used for  the human ovum. These include the zona pellucida
 of stored human  follicular oocytes (53) and the zona-free hamster
 vitellus (54).  Regrettably, these tests  have  not  been  carefully
 validated  to establish variation among independent analyses of the
 same ejaculate or of different ejaculates from one male. In situations
 where in vitro testing of human sperm fertility is indicated, the use
 of a double-fluorescent-label competitive sperm penetration  assay
 with the zona-free hamster egg will increase the sensitivity of the test
 (55). The sensitivity  of the hamster  egg penetration assay is also
 increased by  attempting to count the total number of sperm per
 penetrated  hamster vitellus as well as  the percentage of penetrated
 eggs.

-------
                                85

    Animals. In  vivo  mating  tests  should  be carried out  with
laboratory rats and rabbits. A visible  reduction of quality of the
ejaculate may not be reflected in the  fertility level because of a
superfluidity of spermatozoa in the ejaculate. The sensitivity of the
test can be increased greatly by insemination with critical numbers of
sperm (ca. 1 X 106  to 2 X  106 in the  case of the rabbit). In vitro
tests are unnecessary for use with animals.

Usefulness

    The animal tests are  useful since  they  measure the ability of
sperm to reach and fertilize an ovum. The in vivo tests  are well
established and reliable if critical  sperm numbers are used.  Tests of
the fertilizing capacity of human sperm allow assessment of semen
from a human population at reproductive risk when the hazard being
studied has produced fertilization  dysfunction in the animal tests. If
fully validated, the in vitro  human fertilization system also could be
used  to determine the dose-response relationship of a compound to
the fertility of human sperm.

Sensitivity

    Although simple mating  trials with  evaluation of  offspring
provide some useful information on fertilization, this  is an  all-or-
none measurement.  Since sperm production  is  greatly in excess of
that required for fertility, significant reductions in sperm output by
the testes may not be detectable by this method (2). The test can be
improved  as an  assay of fertilization ability  by using  artificial
insemination with  sperm in limited  numbers. Another potentially
useful approach  involves  competition between  two populations of
spermatozoa from males  whose status in  relation to each  other is
known, with expectation  of  change in the competitive relations
following exposure  of the  male to an agent of interest (39). This
latter approach requires further validation (see Research Needed).
    In fertility tests embryos should be recovered as early as the 2—8
cell stage. Evaluation of a  second group of pregnant rats  between
days 15 and 19  enables comparison  of the  numbers of viable and
dead embryos with the number of corpora lutea and is more  efficient
than allowing parturition.  If rabbit eggs of 2—8 cells are recovered,
the number of sperm associated with fertilized and unfertilized eggs
can be counted.  This should bring to light abnormalities in sperm
transport or abnormalities of cleavage that may result from defects in
the sperm genome (see Sperm Nucleus Integrity).

-------
                                86
Specificity
   The fertilization tests are specific indicators  of reproductive
toxicity, although they have limited value in the broader context of
toxicological testings. The toxicological end points in the in vivo
animal fertilization system include failures of (a) sperm-egg associa-
tion,  (b) sperm penetration of the zona pellucida, and (c) normal
cleavage  of  the  early  embryo.  All  of these  events are  directly
analogous to  those occurring in  humans. A consistent failure of
human  spermatozoa to penetrate >10% of zona-free hamster eggs
probably reflects an abnormality of the physiological events associ-
ated with fertilization (i.e., sperm capacitation and/or the acrosome
reaction). These events  are  presumed to  be  the same  as those
required for human fertilization in vivo.
                     Sperm Nucleus Integrity

    A toxic chemical may cause infertility of exposed males through
action on the sperm genome rather than by alteration of the normal
course of spermatogenesis. Thus, the usual parameters used to assess
semen  quality will not  detect  this cause  of infertility. Genetic
damage to the spermatozoa is best assessed by mating the exposed
male to untreated  females and observing the progeny  for sterility,
heritable  translocations, sex-chromosome loss, specific locus  muta-
tions,  mutations  affecting the  skeleton  and eye,  and dominant
lethality.  These procedures are covered in the U.S.  Environmental
Protection  Agency's proposed  Guidelines  for Mutagenicity  Risk
Assessment (56). Of  these tests,  only dominant lethality  has  a
bearing on the  fertility  of an  exposed male. This  effect can be
detected by evaluation  of fertilized eggs and embryos as outlined in
the section titled Fertility Testing.
    Since the animal tests referred to above are not applicable to the
human male, it would  be desirable to  be able to assess the genetic
integrity  of human spermatozoa  directly.  Four methods for the
detection  of chromosomal abnormalities in spermatozoa  are as
follows:

Quinacrine staining for Y-chromosome aneuploidy
    This  technique, used also by inference  for possible somatic
chromosome  aneuploidy (57),  is  easy and economical,  does not
require sophisticated equipment, and should be suitable for the study
of population groups. However, the  method is subject to many

-------
                                87

errors, and its accuracy has been questioned (58, 59). The reliability
of the procedure can be improved by using more rigid criteria for
scoring fluorescent  Y  bodies  (60).  Nevertheless, the  technique's
reliability has not been sufficiently established to warrant its use as a
routine screening procedure.

Spermatozoal morphology

    Chemicals, radiation, heat,  and a variety of insults increase the
proportion of morphologically  abnormal sperm in the ejaculate, and
an  increase in the  number  of abnormal  sperm usually results in
impaired fertility. This aspect has been considered in the section on
evaluation of semen quality.  That these abnormalities are associated
with chromosomal damage, however, has not been demonstrated.

Karyotyping of human spermatozoa by the
    denuded-hamster-egg technique

    This  procedure is  technically difficult, requires  highly trained
personnel, and at present should be reserved for evaluation of those
cases where additional evidence that chromosome abnormalities are a
factor in reduced fertility is desired.
    Genetic damage with  consequences  for male fertility also can
arise from strand breaks,  base  alterations, and  base  substitution in
the sperm DNA. Except for strand breaks, methods are  currently
unavailable  for  the detection  of these  lesions. Future  work  to
develop qualitative  and quantitative procedures for the detection of
such lesions in sperm DNA would be desirable, since  sperm with an
apparently normal chromosome complement may be responsible for
male fertility problems.
                         Dose Response

    1. The  criteria for evaluating  male reproductive processes, dis-
cussed above, can be quantified. In most cases, the procedures are
objective, accurate,  precise,  and sensitive.  Data for each criterion
have  a considerable response  span,  although values for normal
individuals may not have a normal distribution.
    2. A number of agents acting  on the reproductive system are
known to induce a partial suppression in one or more of the criteria
listed when given in low dosages and a more severe effect as the dose
is increased.

-------
                                  88
    3. It is likely that separate dose-response curves can be estab-
lished for  several criteria with each agent tested. The sensitivity of a
particular test would depend upon the nature of the agent.
    4. It is likely that  agents affecting reproduction have a threshold
dose below which damage does not occur.
    5. It is unlikely, however, that chronic administration of an agent
at >0.5 MTD would  not induce a detectable alteration in  one or
more of the criteria listed, in  at least one of two species, if the agent
in  fact  has  a  deleterious  effect on reproductive function  in the
human male.
    6. Reversibility of damage to the male reproductive system often
occurs after exposure  to the causative agent is terminated. Complete
regeneration  repair  usually will require an interval equivalent to  at
least  three to four  and often more than six to twelve cycles of the
seminiferous epithelium.

                            REFERENCES

 1. Amann, R. P.: Sperm, production rates. In: The Testis, Vol. 1, A. Johnson,
     W. Gomes, and N. Van Demark, Eds., Academic Press, Inc.: New York; pp.
     433-482,1970.
 2. Foote, R. H.: Research techniques to study reproductive physiology in the
     male. In:  Techniques  and Procedures  in Animal Science  Research,
     American Society of Animal Producers: Albany, New York; pp. 81-100,
      1969.
 3. Carson, W. S. and Amann, R. P.: The male rabbit. VI. Effects of ejaculation
      and season on testicular size and function. J. Anim. Sci.  34:302—309,
      1973.
 4. Coulter, G. H. and Foote,  R.  H.:  Bo.vine testicular  measurements  as
      indicators of reproductive performance and their relationship  to produc-
      tive traits in cattle: a review. Theriogenol. 11:297-311, 1979.
 S.Paufler, S. K., Van Vleck, L. D. and Foote, R. H.: Estimation of testicular
      size in live rabbits. Int. J. Fertil. 14:188-191,1969.
 6. Amann, R. P. and Lambiase, J. T.: The  male rabbit. III. Determination of
      daily  sperm production by means of testicular homogenates. J. Anim. Sci.
      28:369-374,1969.
 7. Robb, G. W., Amann, R. P. and Killian, G. J.: Daily sperm production and
      epididymal sperm reserves of pubertal and adult rats. J. Reprod. Fertil. 54:
      103-107,1978.
 S.Johnson, L., Petty, C. S. and Neaves, W. B.: The relationship of biopsy
      evaluations and testicular measurements to overall daily sperm production
      in human testes. Fertil. Steril. 34: 36-40, 1980.
 9. Johnson,  L.,  Petty,  C. S.  and  Neaves, W. B.:  A new  approach  to
      quantification of spermatogenesis  and its application to germinal cell
      attrition during human spermiogenesis. Biol. Reprod. 25: 217-226,  1981.

-------
                                   89

10. Mian, T. A., Suzuki, N., Glenn, H. J., Haynie, T. P. and Meistrich, M. L.:
     Radiation damage  to  mouse  testis  cells from "M technetium  per-
     technetate. J. Nucl. Med. 18: 1116-1122,1977.
11. Amann, R. P.: A critical review  of  methods for evaluation of spermato-
     genesis from seminal characteristics.  J. Androl.; in press, 1981.
12. Lu,  C. C., Meistrich, M. L.  and  Thames, H. O., Jr.: Survival of mouse
     testicular stem cells after gamma or neutron irradiation. Radiat. Res. 81:
     402-415,1980.
13. Meistrich, M. L.,  Hunter, N.,  Suzuki,  N., Trostle, P. K. and Withers, H. R.:
     Gradual  regeneration of  mouse   testicular  stem  cells   after  ionizing
     radiation. Radiat. Res. 74: 349-362,1978,
14.Paufler, S. K.  and Foote, R. H.: Effect of triethylenemelamine (TEM) and
     cadmium chloride  on spermatogenesis in rabbits. J. Reprod.  Fertil. 19:
     309-319,1969.
IS.Swierstra, E. E. and Foote, R. H.: Cytology and kinetics of spermatogenesis
     in the rabbit. J. Reprod. Fertil. 5: 309-322, 1963.
16.Clermont,  Y.: Kinetics  of  spermatogenesis in  mammals:  seminiferous
     epithelial cycles and spermatogonial renewal. Physiol. Rev. 52: 198—236,
     1972.
17. Berndtson, W. E.: Methods for quantifying mammalian spermatogenesis: a
     review. J. Anim. Sci. 44: 818-833,1977.
18. Meistrich, M. L., Reid, B. 0. and Barcellona, W. J.: Changes in sperm nuclei
     during spermiogenesis and  epididymal maturation. Exp.  Cell. Res. 99:
     72-78,1976.
19. Jackson, H.: Comparative effects of some antispermatogenic chemicals. In:
     The  Regulation of Mammalian Reproduction, S.Segel, R. Crozier,  P. A.
     Corfman, and  P.D. Condliffe,  Eds.,  Charles C. Thomas:  Springfield,
     Illinois; pp. 257-270,1973.
20. Oakberg, C. F.: Gamma-ray sansitivity of spermatogonia of the mouse. J.
     Exp. Zool. 134: 343-356, 1957.
21.Parvinen,  L. M.,  Soderstrom, K. 0. and Parvinen, M.: Early effects of
     vinblastine and  vincristine on the rat spermatogenesis: analyses by a new
     transillumination-phase contrast microscopic method. Exp. Pathol. 15(2):
     85-96,1978.
22. Homm, R. E., Rustians, C. and Hahn, D. W.: Antispermatogenic effects of
     5-Thio-D-glucose in male rats. Biol. Reprod. 17(5): 697-700, 1977.
23.Steinberger, E. and  Sud, B. N.: Specific  effect of fluoroacetamide on
     spermiogenesis. Biol. Reprod. 2: 369-375,1970.
24. Lu, C. C. and  Meistrich, M. L.: Cytotoxic effects of chemotherapeutic drugs
     on mouse testis  cells. Cancer Res. 39: 3575-3582,1979.
25. Withers, H. R., Hunter, N. M., Barkley, H. T. and Reid, B.O.: Radiation
     survival and regeneration characteristics of spermatogenic stem cells of
     mouse testis. Radiat. Res. 57(1): 88-103, 1974.
26. Courot, M.: Some results obtained in  the irradiation with X-rays of testes of
     lambs. In: Effects of Ionizing Radiation  on the Reproductive System,
     W.Carlson and  F.Gassner, Eds.,  Pergammon  Press:  New  York: pp.
     279-286,1964.

-------
                                   90

27. Berndtson, W. E.: Methods for quantifying mammalian spermatogenesis: a
     review. J. Anim. Sci. 44: 818-833,1977.
28. Steinberger, E. and Tjioe, D. Y.:  A  method for quantitative analysis of
     human seminiferous epithelium. Fertil. Steril. 19: 960-970, 1968.
29. Harasymowycz, J., Ball, L. and Seidel, G. E., Jr.: Evaluation of bovine
     spermatozoal morphologic features after staining or fixation. Am. J. Vet.
     Res. 37:  1053-1057,1976.
30. Holtz, W. and Foote, R. H.: Composition of rabbit semen and the origin of
     several constituents. Biol. Reprod. 18: 286-292,1978.
31-Amann, R. P.: Use of animal models for detecting specific alterations in
     reproduction. Appl. Fund. Toxicol.; in press, 1982.
32. Boucher, J. H., Foote, R. H. and Kirk, R. W.: The evaluation of semen
     quality in the dog and the effects of frequency of ejaculation upon semen
     quality,  libido,  and  depletion of  sperm reserves. Cornell  Vet. 48(1):
     67-86,1958.
33.Amann,  R.  P.:  Effect  of ejaculation frequency  and breed  on semen
     characteristics  and  sperm  output  of  rabbits. J. Reprod.  Fertil.  11:
     291-293,1966.
34. Freund,  M.: Effect  of frequency of emission on  semen output and an
     estimate  of daily sperm production in man. J. Reprod. Fertil.  6: 269—286,
     1963.
SS.Desjardins, C., Kirton, K. T. and Hafs, H. D.: Sperm output  of rabbits at
     various ejaculation frequencies and their use in the design of experiments.
     J. Reprod. Fertil. 15: 27-32, 1968.
36. Amann, R.P., Kavanaugh, J. F., Griel,  L. C., Jr. and Voglmayr, T. K.: Sperm
     production  of  Holstein  bulls  determined  from testicular spermatid
     reserves,  after  cannulation of rete  testis or vas deferens,  and by daily
     ejaculation. J. Dairy Sci. 57: 93-99,1974.
37.Katz, D. F. and Overstreet, J. W.:  Sperm motility assessment by video-
     micrography. Fertil. Steril. 35(2): 188-193,1981.
38. Overstreet, J. W., Katz, D. F., Hanson, F. W. and Fonseca, J. R.: A simple,
     inexpensive method  for objective assessment of human sperm movement
     characteristics. Fertil. Steril. 31: 162-172, 1979.
39. O'Connor, M. T., Amann, R. P. and Saacke, R. G.: Comparisons of computer
     evaluations of  spermatozoal motility with standard laboratory tests and
     their use for predicting fertility. J. Anim. Sci.; in press, 1981.
40. Seidel, G.  E., Jr. and Foote, R. H.: Variance components of semen criteria
     from bulls ejaculated frequently and their use in experimental design. J.
     Dairy Sci. 56: 399-405,1973.
41. MacLeod, J. and Gold, R. Z.: The male factor in fertility and  infertility. II.
     Spermatozoan  counts in 1000 men of known fertility and in  1000 cases of
     infertile marriage. J. Urol. 66: 436-449,1951.
42. Freund, M.: Standards for the rating of human sperm morphology. Int. J.
     Fertil. 11:97-180,1966.
43.Belsey, M. A., Eliasson, R., Gallegos, A. J., Moghissi, K.S., Paulsen, C. A.
     and Prasad, M. R.N.: Laboratory Manual for the Examination of Human

-------
                                   91
     Semen and Semen-Cervical Mucus Interaction, Press Concern: Singapore;
     43 pp., 1980.
44. Eliasson, R.: Analyses of semen. In: Progress in Infertility, S. J. Behrman
     and  R.W. Kistnes, Eds., Little Brown and Co.:  Boston; pp. 691-713,
     1975.
45. Hammerstedt, R. H.: Monitoring the metabolic rate of germ cells and sperm.
     In:   Reproductive Processes  and  Contraception,  K.W. McKerns, Ed.,
     Plenum Publishing: New York; pp.  353-391, 1981.
46. Thorell, J. I. and Larson, S. M.: Radioimmunoassay and Related Techniques.
     C. V. Mosby Co.: St. Louis; 1978.
47. Gomes, W.  R.:  Chemical agents affecting testicular function and  male
     fertility. In:   The  Testis,  Vol.3,  A.Johnson,  W. Gomes,   and
     N.VanDemark,  Eds., Academic Press, Inc.: New York; pp. 483—554,
     1970.
48. Jouannet, P., Czyglik, F., David, G., Mayaux, M. J., Spira, A., Moscato, M. L.
     and  Schwartz, D.: Study of a group of 484 fertile men. I. Distribution of
     semen characteristics. Inter. J. Androl. 4: 440—449, 1981.
49. Schwartz, D.,  Mayaux, M. J., Spira,  A., Moscato, M.L.,  Jouannet, P.,
     Czyglik, F. and  David, G.:  Study of a group of 484 fertile men. II.
     Relation between age (20—50) and semen characteristics. Inter. J. Androl.
     4: 450-456,1981.
50. Heuchel, E., Schwartz, D. and Price, W.: Within-subject variability and the
     importance of abstinence period  for sperm count, semen volume, and
     pre-freeze and post-thaw motility. Andrologia 13: 479—485, 1981.
5I.Rogers, B. J., Van Campen, H., Veno, M., Lambert, H., Bronson,R. and
     Hale, R.: Relationship between  morphology and human-sperm fertilizing
     ability. Fertil. Stern. 32:  664-670, 1979.
52.Snyder, P. J., Lipschultz, L. I. and Greenberg, S.H.:  In: The Testis of
     Normal and Infertile Men, P. Troen  and H. Nankin, Eds.,  Raven Press:
     New York; 539 pp., 1977.
53. Yanagimachi, R., Lopata, A., Odom, C. B., Bronson, R. A., Malin, C. A. and
     Nicolson,  G.L.: Retention of biologic characteristics of zona pellucida in
     highly concentrated salt  solution—use of salt-stored eggs for assessing the
     fertilizing-capacity of spermatozoa. Fertil. Steril. 31(5): 562—574, 1979.
54. Yanagimachi, R., Yanagimachi, H. and Rogers, B. J.:  Use of zona-free animal
     ova  as  a  test-system for assessment of fertilizing-capacity  of  human
     spermatozoa. Biol. Reprod. 15(4): 471-476, 1976.
55..Blazak, W.  F.,  Overstreet,  J. W., Katz, D. F.  and Hanson, F.W.: A
     competitive in vitro assay of human sperm  fertilizing ability utilizing
     contrasting fluorescent sperm markers. J. Androl; in press, 1982.
56. Federal Register: Proposed guidelines for mutagenicity risks assessments. RP
     FRL 1563-2.45(221): 74984-74988, 1980.
57. Kapp, R. W., Jr.: Detection of aneuploidy in human sperm. Environ. Health
     Perspect. 31:27-31,1979.
58. Beatty, R. A.: F-bodies as Y-chromosome markers in mature sperm heads: a
     quantitative approach. Cytogenet. Cell. Genet. 18: 33-49, 1977.

-------
                                  92
59. Roberts, A.  M. and Goodall, H.: Y-chromosome visibility in quinacrine-
     stained human spermatozoa. Nature 262: 493—494,1976.
60. Kapp, R. W., Jr. and Jacobson, C. B.: Analysis of human spermatozoa for Y
     chromosomal  nondisjunction.  Teratogen.  Carcinogen.  Mutagen.   1:
     193-211,1980.

-------
              II.  GLOSSARY OF TERMS USED IN
                    MALE REPRODUCTION

androgen—a class of steroid hormones produced in the gonads and
adrenal cortex  that regulate  masculine  sexual  characteristics;  a
generic term for agents that encourage the development of or prevent
changes in male sex characteristics.

backward motility—the movement of a sperm in a reverse direction
(toward the middle piece)  rather than a  forward direction.  Note:
backward motility is typically caused  by  a 180° reflection of the
middle piece,  which may be a secondary abnormality or may be an
artifact induced  by temperature shock or osmotic shock.

cellular association or stage—one of a series of characteristic cellular
groupings of different types of germ cells found in a specific area of a
seminiferous tubule. Each association contains several layers of germ
cells, each layer representing one cell generation. These groupings are
not random. Thus, each association contains specific germ  cell types
in certain developmental phases. For example,  spermatogonia of a
specific type  are  always found  with primary  spermatocytes of a
certain developmental phase and spermatids of a certain develop-
mental phase. One cellular association or stage is  found at any
moment in a  given  site  within a tubule. Cellular associations are a
consequence of  the synchronous evolution of the different germ cell
generations.

circular motility—a clearly  discernible motion at a moderate-to-high
velocity, but in circles rather than a more or less linear direction.

cohort of germ  cells—all germ cells that  are the progency of one
A-spermatogonium. Since cytokinesis is incomplete, all germ cells in
the  cohort  remain joined   by  intercellular bridges  and develop
synchronously.

cycle of the seminiferous epithelium—the complete series of cellular
associations occurring in the seminiferous epithelium (6  stages in
man; 14 stages in the rat; and generally classified into 8 stages in the
rabbit).
                                93

-------
                               94

daily spermatozoal output—the total number of sperm  ejaculated
over an interval of at least 7 days after depletion of epididymal
reserves,  expressed on a per-day basis. Note: for males ejaculating
once every  1  to 3 days, after the reserves of spermatozoa in the
cauda epididymidis and ductus deferens have been  stabilized, daily
spermatozoal output will approach daily spermatozoal production.

daily  spermatozoal production—the total number of sperm pro-
duced per day by the two testes.

duration  of spermatogenesis—the interval between the time a stem
spermatogonium  becomes committed  to produce  a cohort  of
spermatids and the release of the resulting spermatozoa from the
germinal  epithelium. It is likely that the duration of  spermatogenesis
requires between 4.3  and 4.7  cycles of the seminiferous epithelium
(exact values  for  most  species are unknown). It  is difficult  to
establish  the time interval between formation of the stem spermato-
gonium and formation of preleptotene primary  spermatocytes, but
this interval may equal the duration of between 1.2 and 1.7 cycles of
the seminiferous epithelium in many species. Therefore, the term
amputated  spermatogenesis is occasionally used to refer  to  the
portion extending from formation of the preleptotene spermatocytes
through  spermiation; this process  typically  requires  about three
cycles  of  the seminiferous  epithelium.  The  entire  duration  of
spermatogenesis would total 4.2 to 4.7 cycles of the  seminiferous
epithelium (about 72  days in the human and fewer in most animals).

duration of the cycle of the seminiferous epithelium—the interval
required for a cell to pass through one complete series  of cellular
associations. This  duration is  constant for a strain  or species (12.9
days for Wistar rat, 10.7 days for rabbit, and 16.0 days for human).
The cycle length is unaffected by environment, hormonal levels, or
cytotoxic damage to the germ  cells.

efficiency   of  spermatozoal  production—the  number  of  sperm
produced per day per gram of  testicular parenchyma.

ejaculate—the total seminal sample obtained during ejaculation.

ejaculation—the expulsion of semen through the urethra.

-------
                                95
emission—deposition  of sperm  and  fluids from  the caudae epi-
didymidis and ductuli deferentia and  fluids from the  accessory sex
glands into the pelvic urethra.

flagellating spermatozoon—a sperm (not stuck to the glass slide)
whose position does not change, although  its tail  moves back and
forth.

follicle-stimulating  hormone  or  FSH—a  glycoprotein hormone
secreted by  the anterior  pituitary of  vertebrates that promotes
spermatogenesis and stimulates growth and secretion of the Graafian
follicle.

luteinizing hormone or LH—glycoprotein hormone secreted by the
adenohypophysis of vertebrates that stimulates hormone production
by interstitial cells of gonads.

maximum tolerated dose (MTD)—the highest dose  that can be given
during a chronic study without a possibility of shortening an animal's
life other than through its carcinogenicity.

meiosis—two  divisions  of  primary  spermatocytes  to first form
secondary spermatocytes and secondly to form spermatids. Cells are
called primary or secondary spermatocytes.

nonmotile spermatozoon—a sperm that does not quiver or move a
discernible distance during visual observation.

percentage of  motile sperm—the  percentage  of  sperm  that  are
progressively motile,  circularly motile,  or backward motile; con-
ventionally  estimated as a  subjective observation of sperm in a
diluted sample of semen viewed with a phase-contrast microscope.
Note:  this percentage can be  determined objectively using  one of
several procedures.

percentage of progressively motile sperm—the percentage  of sperm
that  are  progressively  motile (excluding  circularly motile and
backward motile sperm); conventionally estimated as a subjective
observation of sperm  in a diluted sample of semen viewed with a
phase-contrast microscope. Note: this  percentage can be determined
objectively using one of several procedures.

-------
                               96

primary abnormality—an abnormality of sperm morphology origi-
nating during  spermatogenesis, often  associated  with the  head.
Fertilization  of an ovum by the spermatozoon characterized by a
primary abnormality is unlikely.

progressive motility—a  clearly  discernible, fairly  continuous, for-
ward  motion at a moderate-to-high  velocity in a reasonably  linear
path.  Progressive  motility  is greater  than 25 /mi/sec for human
sperm. Note: in nonfrozen  semen, many sperm will rotate on their
long axis  while swimming progressively, although in frozen-thawed
semen,  progressive motility may  not  be accompanied by cellular
rotation. Also,  the composition of the buffer used to dilute a sample
of semen can influence whether a motile sperm will rotate or swim
without rotation (flat). Rotation about the long axis and the helical
beat of the tail often move the head of the sperm  in a zig-zag path
rather than a true linear path.

quantitative evaluation or objective evaluation—an analytical mea-
surement  of sperm motility or velocity  performed by  a nonbiased
instrument rather than visually by an individual.

quivering  spermatozoon—a sperm that  rotates slightly on its long
axis or oscillates; characteristic  of some sperm recovered from  the
efferent ducts or rete testis.

secondary abnormality—an  abnormality  of  sperm  morphology
induced during epididymal  transit or ejaculation, usually associated
with  the tail.  When a  secondary  abnormality is induced in a
spermatozoon,  its competitive ability to fertilize an ovum is reduced.

semen—a mixture of sperm and fluids from the excurrent ducts and
accessory sex glands.

seminal volume—the volume  of an ejaculate  (expressed in milli-
liters).

seminiferous epithelium—the  normal  cellular components within
the seminiferous tubule consisting  of  Sertoli cells and germ cells
(spermatogonia, primary spermatocytes, secondary spermatocytes,
and spermatids).  Sertoli cells are  somatic  cells that are usually
nondividing  in adult  animals and probably are important  for
metabolic exchange between the germ cells in the luminal compart-
ment  and that,  by  means of Sertoli-Sertoli junctions, form  the

-------
                                97
blood-testis barrier. They also aid in coordination of spermatogenesis
and have an endocrine function.

spermatogenesis—the sum of  the transformations  that  result in
formation  of  spermatozoa  from  spermatogonia  and continued
formation  of a fairly constant number of uncommitted spermato-
gonia.  The  entire  spermatogenic  process is  initiated  in early
embryonic development and  continues after birth and puberty as a
consequence of continual renewal of stem cells. At birth two  cell
types are found within the  seminiferous tubule:  supporting cells,
which give  rise to the Sertoli  cells  of the puberal male, and  the
gonocytes,  which  will  develop into spennatogonia. The  intense
proliferation of germ cells and the subsequent release of spermatozoa
do not  occur randomly. Rather the germinal elements always follow
the same pattern  of  development (unless particular cells and their
progeny degenerate) within males of a species.

spermatozoal velocity—the  velocity with which  a progressively
motile  or circularly motile sperm  moves. Spermatozoal velocity is
conventionally expressed on a subjective scale from 0 (low velocity)
to 4 (maximum velocity) but  should  be expressed as Mm/sec  on  the
basis of quantitative measurements.

spermiation—release  of spermatozoa from the germinal epithelium
into the lumen of the seminiferous tubule. Prior  to release the germ
cells are called spermatids, and after spermiation they are called
spermatozoa.

spermiogenesis—the  differentiation  of  spermatids  from spherical
cells with considerable  cytoplasm to characteristically shaped cells
with a  highly  condensed nucleus and scant cytoplasm but  with a
flagellum. Cells are called spermatids.  Based on changes  in  the
spermatid  acrosome,  spermiogenesis  can  be  considered   as a
continuum consisting  of four  phases: Golgi, cap,  acrosome, and
maturation. In addition to acrosomal evolution, condensation of the
nuclear material and formation of the flagellum occur.

subjective evaluation—a visual estimate subject to observer bias and
error.

testosterone—a biologically potent androgenic steroid that may be
released from the gonads and adrenal glands.

-------
                               98

total spermatozoa per ejaculate—the total number of spermatozoa
in an ejaculate (determined as the product of seminal volume times
spermatozoal concentration and  expressed as 106). Note: the total
number of  sperm per ejaculate, not spermatozoal concentration,
provides the best information  on  the  number of  spermatozoa
produced  by the  testes,  since  spermatozoal concentration is  in-
fluenced by the relative contributions  of the accessory sex glands
diluting the  bolus(es) of sperm transported during emission from the
ductus deferens and cauda epididymidis.

twitching spermatozoon—a sperm that occasionally or continuously
moves a short distance with a violent motion and then comes to rest,
at least momentarily, before  an  additional twitch or jump. The
twitch or jump need not be in a forward direction.

-------
                        CHAPTER 4
  CURRENT STATUS OF, AND CONSIDERATIONS

   FOR, ESTIMATION OF RISK TO THE HUMAN

       CONCEPTUS FROM ENVIRONMENTAL

                      CHEMICALS


                    Definition and Scope

   Teratology is the study of the causes,  mechanisms, and sequelae
of perturbed developmental events in species of animals that undergo
ontogenesis.  This report is restricted to a consideration of factors
influencing the current status of risk assessment of teratologic effects
of environmental agents. It is considered a preliminary document
touching  upon the  major  considerations basic  to quantitative
estimation of risk to development of  the  conceptus  following
exposure  of  pregnant animals to environmental agents. This docu-
ment provides no definitive means for  assessing risks to the human
conceptus, since  no  documented  or  validated  system  for  such
assessment has yet been established. Basic to risk estimation is hazard
assessment, which requires quantification and validation of reliable
end point assays. This document briefly discusses the factors and
scientific considerations upon which degrees of confidence applicable
to contemporary  studies  of  teratology   are  to be based.  Some
additional considerations in evaluating experimental data (e.g., acute
versus chronic exposures) have not been covered explicitly, but
references are provided  to aid the  reader in gathering further
information.


       Impact of Developmental Abnormalities on Humans

   Approximately 50% of human conceptuses fail to reach term,
and perhaps  as many as half of those lost are structurally  abnormal
(1). Approximately 3% of newborn children are found to have one or
more significant congenital malformations at birth, and by the end of
tiie first postnatal year, approximately 3% more (2, 3) are found to

                             99

-------
                               100
have developmental malformations. An additional group, whose size
is difficult to estimate, has functional abnormalities of the nervous,
respiratory, gastrointestinal, immunologic, and other systems. Some
unknown proportion of these abnormalities may be due to environ-
mental insult during prenatal life.

               Causes of Congenital Malformations

    Relatively  little  is known  about  the specific  causes of most
human  congenital defects. It is estimated that 10 to  15%  of  all
human  congenital malformations are due to environmental agents
and another 10 to 15% to hereditary factors (i.e., gene mutations and
chromosomal  aberrations). The remainder are  considered to result
from unknown causes and from  complex interactions between
multifactorially  determined hereditary susceptibilities  and  micro-
environmental  factors  precipitating  abnormal developmental  se-
quences within the conceptus and its associated membranes. To date,
only a relatively small number  of specific environmental agents and
factors have been identified as causing human malformations (4).
    From the above, it is concluded that although regulatory controls
on  man-made environmental agents may reduce the incidence  of
developmental abnormalities, they will not totally prevent them. It
must be  recognized that indications from  animal experiments  of
adverse effects of environmental agents on development may not
always be corroborated by observations of perturbed development in
human  populations. Nevertheless,  and in full  recognition of these
qualifiers, standard animal testing is  presently considered the best
available method for predicting risk of congenital malformation in'
human  beings prior to human  exposure. Information derived from
such testing can be used to detect and to estimate the magnitude of
hazard posed by specific substances to human prenatal development
and can serve as a basis for estimation of risk.

              Qualitative Evaluation of Risk Potential

Interspecies comparisons
    Inherent interspecies differences  complicate  extrapolation  of
animal  test results to direct determination of human risk. Because a
species  identical to the human  in all relevant characteristics does not
exist, interspecies differences  between human beings and the test

-------
                                101

species must be considered when data are being evaluated. Interpreta-
tion of  these  inherent  interspecies differences  is complicated by
species differences in metabolism and pharmacokinetics of the test
agent and in developmental and other attributes characteristic of the
species. Very little  is currently  understood  about the extent and
nature of the interplay among these many factors as they may affect
the production of a teratogenic event.
    Since human beings  are manifestly heterogeneous, there is little
doubt that  human populations  will  contain  broad  degrees of
susceptibility and resistance to the possible adverse prenatal effects
of  environmental  agents.  Because  this heterogeneity  is  largely
determined by genetic variability, it has been reasoned that stocks of
animals bred at random  are the most appropriate models for testing
teratogenicity.  However,  in order  to  estimate  the  degrees of
susceptibility that may  exist within human  populations, both the
average response  of the test group and the extent of responses within
it must be considered. This goal can be achieved to some extent by
using several stocks  of animals. To make such a procedure even more
sensitive  and useful, several inbred strains may also be tested, since
this procedure increases the likelihood that a range  of sensitivities
will be uncovered (5). For instance, genetically controlled variations
in embryonic  face  formation account  partly for the sensitivity of
certain mouse strains to spontaneous  (6)  and teratogen-induced
(7, 8) cleft lip and isolated cleft palate.

Dosing and mode of administration

    The  test agent  should  be administered over a range of doses,
including a level sufficient  to produce  signs of maternal toxicity in
the  particular species used. If a teratogenic response is observed, a
dose-response relationship should be determined for the agent and
that specific teratogenic effect. In using test animals, the selection of
dosing intervals  must take into  account  the varying degrees of
sensitivity during organogenesis  in that species, the possibility of
enzyme  induction  or other modifying  processes  that could result
from repeated administration of the test material,  and the practical
aspects of administration that would make the dosing comparable to
that which would likely occur with human exposure.
    The  route  of administration of the test agent  may significantly
affect the outcome of  an experiment. In  general, the route of
exposure for test animals  should  mimic that of  human exposure
where possible, although valuable data may be obtained from other
routes of exposure as well. Differences in the response of a species to

-------
                                102

the route, dose, and vehicle used for exposure to the test agent may
result in significant variations in blood and tissue levels of the agent
in the maternal and embryo-fetal units (9). These factors may or may
not  be of direct significance  to teratogenesis, but they must be
recognized as being potentially significant.

Placental transfer
    The anatomy and physiology of the placentas of experimental
animals  and  man  present  a  diverse  spectrum  of maternal-fetal
connections  (10).  The  chorioallantoic  placenta  of the human is
approximated by that in  some  non-human primates,  whereas the
common experimental animals have, in addition, a yolk sac placenta,
which also structurally and  functionally joins embryo and mother.
The extent  to which the yolk sac may  supplement or complement
transfer  of  a previously untested chemical  via the  chorioallantoic
circulation is largely unpredictable. In most of these species (e.g., rat,
mouse, rabbit, guinea pig), the yolk sac placenta may play a major
role in maternal-fetal exchange  of substances during early  organo-
genesis. The chorioallantoic  placenta,  which is readily available for
convenient  study  at term, is  in  most cases a  totally different
structure from  that  effecting  transfer during the critical stages of
development; therefore great care must  be exercised  to avoid
unwarranted  extrapolation  from  studies of term  chorioallantoic
placenta  to  presumptions for the function of the two placenta!
structures present earlier in gestation.  Lipid solubility, ionic charge,
molecular size,  and  specific structural  configuration all appear to
contribute to the transfer of chemicals between  mother and fetus.
Little  or no relationship may exist between the embryonic and fetal
concentration of agents and their possible teratologic effect, since
potent teratogens  do  not  always  accumulate in the fetus  at
concentrations greater than those of agents with  low teratogenic
potential. Consequently, increased concentration ratios between the
conceptus and  mother do  not  necessarily  allow predictability of
teratogenic   or  other  embryotoxic  potential  (11, 12). Little  is
currently known about the sites of  action  of  teratogenic agents;
therefore, any component of the entire  maternal-placental—embryo-
fetal unit and  all combinations of such  should be considered  as
possible  site(s)  for  teratogenic  action, until the  mechanism of
teratogenic action for given agents is better understood.

-------
                                103

 Pharmacokinetics and metabolism

    Other variables that may affect the teratogenicity of an agent in
 various species include pharmacokinetics and metabolism and those
 exogenous factors  that may  affect these parameters. Some of the
 factors to be considered  when  evaluating data from these experi-
 ments  are  seasonal and  circadian  effects  on development  and
 metabolism (13, 14); interaction of pharmacokinetic and placental
 hemodynamics (15); possible sites of  action  of agents;  sites of
 maternal, placental, and embryonic-fetal metabolism; and deposition
 and/or depression or induction of metabolic enzymes (9, 16). Certain
 agents  may  stimulate  or  inhibit  enzyme  systems, such  as  liver
 microsomal enzyme systems.  In some cases single doses at critical
 periods  in  gestation  induce  a  greater  teratologic  response  than
 divided doses on several consecutive  days (17-21). Maternal  and
 embryo-fetal nutritional and endocrine states in various species may
 interact with and/or alter metabolism and pharmacokinetics (22), as
 may species-specific effects resulting from repeated administration of
 the test agent, saturation of metabolizing enzymes, and inhibition or
 induction of biotransforming enzymes (16, 23).
    Basic  to  interpreting  data from studies of teratology is docu-
 mentation of a dose-response relationship and determination of a
 treatment level below which adverse effects are not evident in  the
 data available (no-observed-effect level or NOEL). Threshold levels
 may  be  encountered,  and  dose  levels  can exist below  which
 development of  the conceptus  suffers  no  observable  deleterious
 effect  at  term.  The no-observed-effect  level does  not  guarantee
 absolute  safety,  because uncertainty  may  result from  biological
 and/or statistical  variation. Failure to detect a deleterious effect on
 the end point examined could indicate the absence of a deleterious
 effect, but  absence of observed effect also could occur  if  the
 magnitude of an  effect were below the limit of statistical detection
 ability.

Mechanisms of action

   The mechanisms underlying  abnormal embryonic development
 are not well understood. It has proven difficult to determine whether
 an observed incidence of abnormal development is the result of an
 agent or one of its products acting directly on the conceptus or its
placenta, or if it is achieved indirectly through an initial effect on the
mother. Therefore,  the primary site of action by an agent capable of
 disrupting development may or may not  be the specific malformed
organ and may not  even be within the conceptus. Whether or not a

-------
                                104

chemical is evenly distributed within the mother and conceptus, its
action on a particular tissue or organ may be dependent on cellular
interactions and the particular developmental events characteristic of
specific ontogenetic stages. Increased knowledge of these ontogenetic
events and the  interaction  of toxic agents or their products  with
them is needed  both to understand the resulting  defects and to
enable  better  extrapolation  of  effects  seen  in one  species to
predictions of potential effects in another.
                         Animal Studies

 Standard teratogenicity testing
    Schardein (24) has discussed in some detail the current method-
 ology and testing approach initially outlined in the 1966 Food',and
 Drug Administration's (FDA's)  guidelines  for reproductive studies
 and in the 1967 and 1978 World Health Organization's recommenda-
 tions, which  are further  specified  in  the  U.S.  Environmental
 Protection Agency's  proposed guidelines. Numerous countries have
 required studies that are generally similar but that vary in particulars.
 The  object  of the standard protocols is to expose animals to test
 materials before breeding of the parental generation, during in utero
 development and lactation,  and in some instances into adult life of
 the offspring.  To achieve their goals, the experiments are designed in
 three phases or segments, with a multigeneration test for reproduc-
 tive effects required in some instances.
    The first phase,  or Segment-I protocol, calls for dosing of both
 male and female  animals to begin  some  calculated time prior to
 breeding.  Treatment  of the young  males begins  60 days prior to
 breeding,  and exposure  of the females to the test substance begins
 two weeks prior to breeding. Dosing continues for both sexes during
 the  breeding  interval and for the impregnated  females throughout
 pregnancy  and lactation. Other  major details of the  Segment-I
 protocol  could be  described  here,  but these  will be  slighted to
 emphasize the basics.
     The Segment-I  protocol is supposed to  examine for possible
 adverse effects on  estrus;  sexual  performance;  formation  of the
 gametes; their release from the gonad, transport, and interaction to
 form a zygote; and zygote passage to and  implantation into the
 decidua.  Because dosing of the dam continues  after mating, the
 protocol could reveal effects on placenta! formation and its function;
 and because dosing continues throughout gestation and lactation, the
 protocol could reveal adverse effects on embryonic or fetal develop-

-------
                                105

 ment and on delivery, nurture, and postnatal development of the
 pups.  Adverse  effects  of  the test material  on  the  supportive
 functional parameters essential for normal occurrence of each of the
 above could also become evident. These effects could be as diverse as
 effects on food intake or altered  endocrine status. .This study is
 usually made in rats, and by indicating problem areas, it can serve as
 a preliminary to later studies.
    The second protocol is  oriented  more specifically  to detect
 effects on embryonic  development. The study is usually made in
 both rats and rabbits. The Segment-II study requires that the males
 not be treated  with the test compound  and that treatment of the
 pregnant females not begin until after decidual implantation of the
 blastocysts  has. occurred. Treatment ceases  at  the end of major
 organogenesis,  usually considered  as the  time  of  closure  of  the
 secondary palate in the species. Autopsy is performed the day before
 expected delivery,  when the  term fetuses  are  collected for gross
 external examination,  after  which  they  are examined for skeletal
 development and internal soft-tissue morphology.
    The goal of this experiment is to detect adverse effects of a test
 material on the developmental events characteristic of major organo-
 genesis in the embryo.
    A Segment-Ill  evaluation  is  a perinatal  and postnatal study
 requiring treatment of the dams only. The test agent is administered
 during  the  last  third  of pregnancy and  throughout  lactation.
 Treatment is  not scheduled to begin until after the period of major
 embryonic development is completed.
    The Segment-Ill safety evaluation was designed to detect adverse
 effects  of  substances  on fetal development  as  well  as those
 developmental processes that continue into infancy and adolescence.
 It is in  this study that potential effects on postnatal behavior of the
 young are usually evaluated.
    A rather  elaborate  multigeneration  protocol is employed for
 evaluating selected substances for effects  on reproduction over three
 generations. The goal of the multigeneration protocol is to reveal
 effects caused by accumulated toxicity or by agents effective at low
 concentration.  These protocols for  safety  evaluation  have  had
 detailed discussion (14, 25), and for each  a data base of considerable
 size has accumulated. They  are not considered  as final,  however,
 because  there is  a need for flexibility  and exercise  of  scientific
judgment (26),  which could improve their detecting ability in some
 instances. There is also some justification for their revision in light of
 research findings in related fields since their inception.

-------
                               106

   There are four types of developmental defects: gross anatomical,
death  in  utero,  growth retardation,  and functional deficit (4).
Currently, the first tliree of these end points are the only ones that
have a data base sufficient to ensure confidence in their applicability
for use in regulatory decisions. Functional status has been studied
broadly only in recent years and soon may develop end point assays
with specific applicability.
    Examination of fetuses to identify gross anatomical defects often
entails judgmental and subjective appraisals based  on criteria or
standards established by individual laboratories. The  routine test as
performed by many laboratories applying FDA's Good Laboratory
Practices requires highly trained technical and professional personnel.
Even though general standards for defining  the limits of normality
and associated terminology have  not  been  established, in  general
when  selected compounds have been  evaluated by various labora-
tories, similar findings  have been  demonstrated  by those  using the
routine teratology test and its methods  for examining the young
(27, 28).
    The  overwhelming  majority of chemicals known to be terato-
genic in human beings  have been demonstrated to be teratogenic in
one or more common  laboratory  species.  Many other agents shown
to be teratogenic  in laboratory animals  have  not yet been docu-
mented  as teratogens  in  humans. The difference may be due to
insufficient epidemiologic data, dissimilarities of exposure levels, or
differences in end points analyzed.  A teratogenic response in one
species or strain  should  be  considered  indicative  of a potential
teratogenic hazard for  human  beings. However, negative responses in
a few species of experimental animals do not necessarily  guarantee
 absence of adverse effects in human conceptuses.  In  the routine
 teratology test, no one species has been consistently more predictive.
 of human teratogenicity than another species.

 Functional teratogenicity testing
     Functional  alterations may prove to be sensitive indicators of
 teratogenic potential. Among those that have been studied following
 prenatal  exposure, a broad and complex range of behavioral effects
 has been described. There is concern that these effects may occur at
 doses below those producing gross structural  defects or prenatal
 death (29). Current literature  is based largely on studies of rodents,
 particularly rats, in which it has been demonstrated that exposure to
 chemicals during  periods from early  organogenesis  through pubes-
 cence can result in behavioral impairments. To detect such effects

-------
                                107

 more  efficiently, reliable  and sensitive test procedures are being
 developed in several  laboratories  (30, 31). Although there are no
 agreed-upon testing methods, current studies routinely  include the
 end points in the following areas: (a) reflex ontogeny, (b) habitation
 and reactivity, (c) learning and problem solving, (d) activity level, (e)
 motor skills, and (f) sensory processes.
    Other functional  parameters  demonstrated  to be affected by
 prenatal  exposure to chemicals include fertility; reproduction; the
 endocrine system; immune competence; xenobiotic metabolism; and
 various  physiologic  parameters,  including   cardiovascular,  renal,
 gastrointestinal, respiratory, and hepatic functions (32). Finally, late
 sequelae  of prenatal  exposure to  chemicals may  be  manifested
 postnatally as cancers  or shortened life span (33, 34).
    Gross  structural defects  or significant growth retardation may
 complicate analysis of data from tests of function, and  alertness to
 potential confounding factors is  essential.  Permanent  changes in
 functional systems should  be viewed as indicating the potential for
 an adverse effect in human beings.  Transient  changes or delays in
 functional ontogeny are  still not understood, and  their significance
 must be further evaluated.
                  Short-Term Testing Procedures

Prioritizing of chemicals for in-depth study

    As a prelude to estimation of potential risks, a series of biological
and informational  factors may  be applied to a new substance to
possibly trigger further testing to some  level in a tier system of
evaluations for teratologic effects. It is considered highly desirable
that substances be prioritized for testing to focus research attention
more readily on substances injuring conceptuses at doses significantly
below  those  toxic  to adults.  In  attempting to list the factors to be
taken  into consideration, the need for short-term systems became
evident because of the large number of evaluations needed and the
fact that many of the available data would be in category 2 below.
Listed  below are the factors  that, when applicable, would make a
substance a high-priority candidate for further testing. Within each of
the  two categories, the factors are  listed in decreasing  order of
importance.

1. Biological  effects data possibly available regarding a substance:
   Suspected  human   teratogenicity;  Teratogenicity  in domestic

-------
                                108

   animals or wildlife established; Short-term teratology test indi-
   cating a significant developmental hazard potential; Adult toxic
   dose/developmental toxic dose ratio large; Toxicity documented
   in the adult at low dosage;
2. Additional information  available  regarding  a substance:  Large
   numbers of  women  exposed; Bioaccumulation  evident; Persis-
   tence of substance in environment; New substance; Involuntary
   exposure.
   The number of agents in use and potentially impinging on human
development is already vast and is increasing rapidly.  Some unknown
small fraction  of  these may be potentially  harmful. to human
conceptuses at doses  below those obviously deleterious to adults.
Short-duration  and low-cost methods for detecting  and prioritizing
those  substances  posing  the greatest potential  hazard  to  the
conceptus  are needed. Because standard tests in animals are quite
costly,  only a  rather  small  number of substances of potential
teratogenic risk can be evaluated each year. This situation requires
development and validation  of short-term methods  that will permit
rapid and meaningful testing of  these chemicals. It is necessary to
develop, validate,  and use assays that will permit more economical
testing of a larger  number of agents than could be tested rapidly and
conveniently  by  standard  teratogenicity  evaluations.   Validation
should consist  of  various forms  of positive correlation between the
results of  such  tests and  those  found in conventional in vivo test
procedures. Particular attention  should be directed to correlations
with known human' teratogenic responses whenever  reliable data are
available. Included  should be chemicals  already known  to have
significant hazard  potential for the conceptus, as well as chemicals
considered as lacking such potential. If properly validated short-term
tests were  to indicate  either  potential  hazard  or safety,  such
determinations would be helpful for establishing a priority system
for further tests aimed toward quantitative risk assessment.
Characteristics of short-term assays
    Short-term tests might be  considered as either preliminaries to
more detailed evaluations, or they might serve as efficient means to
detect those substances capable of posing the greatest hazard to the
conceptus.  Whether  they serve  either or both  of  these  slightly
different  goals, the  tests should possess  certain attributes:  they
should be rapid, economical, and reproducible from one laboratory
to  another;  should have easily identifiable end points; and ideally

-------
                                109

should prioritize substances according to their potential for posing
hazards to the conceptus. Confidence in their applicability would be
increased  by demonstration of  dose-response  relationships. They
should  give  minimal  false negatives; it  is  understood that  false
positives can be explored further in more elaborate animal testing.
Ideally, the system  should encompass as many as possible of the
developmental events known to occur in the conceptus.
    Short-term tests  may serve as preliminary screens to aid in the
detection  of possibly teratogenic hazards. To accomplish this, several
such tests would  probably have to be performed concurrently or
sequentially.  It  must  be remembered  that such indications  of
teratogenic hazard potential must be used prudently for estimating
risk to human beings. Risk estimation can only be achieved by the
use of systems that have been extensively validated, and to date, only
the more  routine standard testing methods are considered applicable
to this use.
Potential short-term systems

   In  vivo. Two  in  vivo systems  have been advanced as possible
short-term systems. One is  an abbreviated version  of a standard
teratology test protocol using the maternal maximum tolerated dose
and  neonatal  evaluation  shortly  after birth (35). The second is
evaluation of homeotic shifts that may prove effective for detecting
minimal expression of teratogenic hazard potential (36). Each system
has potential merit, but as in the in vitro systems, needs careful peer
review or detailed validation.
   In  vitro.  Artificial invertebrate "embryos," embryonic insect
cells, amphibian and fish embryos, or organs of avian and mammalian
embryos (palatal shelves, tooth bud, kidney mesenchyme, pancreas,
bone primordia, lens, sex organs, etc.)  have the potential to serve as
the basis for in vitro systems. Table 12 lists a number of potential in
vitro systems.  In these procedures, cells,  organs, or whole embryos
have been exposed to various chemicals and their effects measured
with the end point permitted by each  system. When attempted in a
few instances,  adequate dose-response relationships were obtained by
some systems. In  their present state, the  systems listed in Table 12
have  been  only  partially  validated   (36-50),  and  their closer
examination is necessary.

-------
                               110
TABLE 12 Some/n Vitro Short-Term Systems
Currently in Various Stages of Development
System
Invertebrates






Fish



Vertebrate cell
culture







Organ culture


Whole embryo culture


Developmental
parameters
monitored References
Hydra
Planaiia




Drosophila cells
Zebra fish



Chick embryo
neural crest
Chick embryo
limb bud
mesenchyme
Mouse tumor cells
Terato carcinoma
stem cells

Mouse embryo
limb bud

Rat, mouse,
chick

Various
Regeneration,
dose-response
relationship
of developmental
toxicity
Differentiation
Dose-response
relationship of
developmental
toxicity
Morphology,
differentiation
Differentiation


Cell attachment
Differentiation
(in vivo or
in vitro)
Growth, dysmo'rpho-
genesis,
differentiation
Growth, dysmorpho-
genesis,
histogenesis
36
37,38




39
40



41

41


42
43


44,45
46,47

16,48
49,50

                  Quantitative Risk Assessment

   Quantitative risk assessment is based on the relationship between
laboratory  findings  and  expected human  response.  If an  agent
demonstrates teratogenicity in any mammalian species, some concern
about prenatal human exposure to the agent is justified. The level of
concern is to be tempered by numerous considerations, not the least
of which is the extent of maternal toxicity evident at the dose level
needed to elicit a toxic response in the conceptus (51).  It is assumed
that margins of safety applied to the experimental data in test species
can be used to estimate an allowable exposure in pregnant women. It
is  considered that no-observable-effect and/or  threshold  levels of

-------
                                Ill

exposure do exist (52) for at least some teratogens. The determina-
tion  of human  risk requires  the definition  of moderating (or
modulating)  conditions such as  the distribution of the compound
within the environment; its  pattern of use and exposure  (whether
intermittent  or chronic); and the identification of those subpopula-
tions that may be at high risk as a result of factors such as  life style,
age, occupation, etc.
    The use of animal test systems  under highly controlled experi-
mental conditions has conditional validity for defining human risk.
Although only warning systems at best, laboratory  experiments can
provide,  in addition to the factors already mentioned, two types of
information  that  may  be useful for estimation of  the  potential
human risk.  These are (a) the ratio  of the adult and developmental
toxic doses  and (b) the shape  of the curve of the  teratogenic dose
response. Although not markedly informative to date, more detailed
delineation  of projected effects may  be obtained  through use of
pharmacokinetic  information, focusing on access of  the  agent  to
relevant  site(s) of teratogenic action (53). However, knowledge of
teratogenic  mechanisms and identification  of  the sites  actually
relevant must be obtained before these considerations can achieve
their full potential utility.
    It is often necessary to  conduct animal experiments at dosage
levels exceeding estimated levels  of human exposure to increase the
likelihood that a weak teratogen  will produce an apparent effect and
to compensate for the relatively small numbers of animals used in the
test. This requires extrapolation of results from experimental dosage
levels to lower levels of human exposure. There is no uniform  basis
for selecting  the appropriate  mathematical model for such extrapola-
tion.
    Safety  factors may be applied  to  establish  acceptable  dosage
levels that are expected to yield acceptable levels of risk. The size of
the  safety factor depends upon the quality and  quantity  of the
biological effects data available.
    For many biological systems, the dose-response curve  tends  to
flatten at low doses,  and for some  teratogens this is  an important
consideration (54). Hence, decreasing dosage by a safety factor of F
will generally decrease risk by more than a factor of F. That is, if the
upper confidence limit on the risk is U at an experimental dosage of
d, the potential risk at a lower  dosage of d/F  is predicted to be less
than U/F for the test animal. The uniformity with which this would
apply to  potential teratogenic  hazards is undetermined as is the
degree of interspecies uniformity for the difference  between the
adult (A) toxic  and developmental (D)  toxic doses. Presence  or

-------
                               112

absence of uniformity in the A/D ratio or slope of the dose-response
between studies in  different species  would also influence the
magnitude  of safety factors.


            Priorities for Future Research in Teratology

    The areas of research in teratology recommended below focus on
two broad objectives: (a) development of practical and informative
testing  systems  with which  to  evaluate  both  the  plethora of
chemicals now in existence and those yet to be developed and (b)
scientific advancement in teratology so that the currently employed
and largely standard test systems  can  become more  useful and
reliable for human risk estimation.  The second objective  does not
imply  that currently  available  methods cannot be used  for human
risk estimation. An opposite view is held,  and within limits, such
estimations are possible at  the present time on the basis of data from
current state-of-the-art studies. It is  felt, however, that methods are
needed to identify more rapidly those chemicals potentially the most
hazardous  and to expand the understanding and applicability of all
test methods.
    1. The  degree to which the end point determinations of adverse
effects  on development encountered  in the  standard protocols
predict adverse effects in other species (especially humans) has not
been reported in detail sufficient for precise quantification of human
risk. Such  studies are encouraged as are those that may indicate how
the predictive ability of tests in animals can more precisely herald
human responses.
    2. Validated  methods  are needed  for  rapid and inexpensive
detection  of  substances uniquely  toxic  to  conceptuses (i.e., sub-
stances that are  embryotoxic at doses below those producing adult
toxicity).
    3. A better understanding of mechanisms of teratogenic action or
elucidation of the steps in the pathway between exposure and effect
might  significantly improve and  refine  end point assays. Similar
studies of pathogenesis are needed  for effects on biochemical and
physiological systems in the dam, uterus, and placenta.
    4. Development of a broader data base on comparative metabo-
lism and pharmacokinetics correlated with  teratologic  end points
may eventually enhance ability to make interspecies extrapolations.
    5. There is a  need to develop and validate methods to detect and
quantify possible functional impairments.
    6. Methods  are  needed  to  detect  and  predict additive  or
synergistic effects more effectively.

-------
                            REFERENCES

 l.Hertig, A. T.:  The overall problem  in man. In:  Comparative Aspects of
     Reproductive Failure, K. Benirschke, Ed., Springer-Verlag: New York; pp.
     11-41,1967.
 2. McKeown, T. and Record, R. C.: Malformations in a population observed for
     five years  after birth. In:  Ciba  Foundation Symposium on Congenital
     Malformations, G.E.W. Welstenholme  and C.M. O'Conner, Eds., Little
     Brown: Boston; pp. 2—16,1963.
 S.Mellon, G. W. and Kalzenstein, M.: Increased incidence of malformations —
     chance or change? J. Am. Med. Assoc. 187: 570-573,1964.
 4. Wilson, J. G.: Environment and Birth Defects. Academic Press: New York;
     305 pp., 1973.
 S.Kalter, H.:  Interplay  of intrinsic and  extrinsic  factors. In: Teratology:
     Principles and Procedures, J. G. Wilson and J. Warkany, Eds., University of
     Chicago Press: Chicago; pp. 57-80, 1965.
 6.Trasler, D. G.: Pathogenesis  of cleft lip and its relation to embryonic face
     shape in A/J and C57BL mice. Teratology 1: 33-49, 1968.
 7.Trasler, D. G.: Aspirin-induced cleft lip and other malformations in mice.
     Lancet 1: 606-607, 1965.
 S.Trasler, D. G. and Fraser, F.  C.: Time-position relationships with particular
     reference to cleft lip and cleft palate. In: Handbook of Teratology, Vol. 2,
     J.G. Wilson  and  F.C. Fraser,  Eds., Plenum  Press:  New York; pp.
     271-292, 1977.
 9. Larsson, K. S.: Contributions of teratology to fetal pharmacology. In: Fetal
     Pharmacology, L. O. Boreus, Ed., Raven Press: New York; pp. 401—418,
     1973.
10. Beck, F.: Comparative placental morphology and function. Environ. Health
     Perspect. 18: 5-12,1976.
ll.Waddell, W. J. and Marlowe, G. C.: Disposition of drugs in  the fetus. In:
     Perinatal Pharmacology and Therapeutics,  B. L. Mirkin, Ed., Academic
     Press: New York; pp. 119-268,1976.
12. Wilson,  J. G.,  Scott, W. J., Ritter, E.  J. and Fradkin, R.:  Comparative
     distribution and embryotoxicity  of methotrexate in pregnant rats and
     rhesus monkeys. Teratology 19: 71-80,1979.
13.Barr, M., Jr.: Prenatal growth of Wistar  rats: circadian periodicity of fetal
     growth late in gestation. Teratology 7: 283-288, 1973.
14. Layton, W. M.:  An analysis of teratogenic testing procedures. In: Congenital
     Defects, New Directions in Research,  D. T. Janerich, R. G. Skalko and
     I. H. Porter, Eds., Academic Press: New York; pp. 205-217,1974.
15.Wolkowski-Tyl, R. M.: Strain and tissue differences in  cadmium-binding
     protein  in cadmium-treated mice. In: Developmental Toxicology of
     Energy-related Pollutants, D. D.  Mahlum, M. R. Sikov, P. L. Hackett and
     F. D. Andrew, Eds., Technical Information Center: Oak Ridge, Tennessee;
     pp.568-585,1978.

-------
                                   114

16.Fantel, A.  G.,  Greenaway,  J.  C., Juchau, M. R. and  Shepard,  T. H.:
     Teratogenic bioactivation of cyclophosphamide in vitro. Life  Sci. 25:
     67-72,1979.
17. Russell, L. B., Badgett, S. K. and Saylors, C. L.: Comparison of the effects of
     acute, continuous,  and fractionated irradiation during embryonic devel-
     opment. Int. J.Radia. Biol.,Suppl: 343-359, 1960.
18. Wilson, J. G.: Effects of acute and chronic treatment with  Actinomycin D
     on pregnancy  and the fetus in the rat. Harper Hosp. Bull. 24: 109—118,
     1966.
19. King, C. T. G., Horigan, E. and Wilk, A. K.: Fetal outcome from prolonged
     versus acute drug administration in the pregnant rat. In:  Drugs and Fetal
     Development,  M. A. Klingberg, A. A. Abramovici and J. Chemke, Eds.,
     Plenum Press: New York; pp. 61-75,1972.
20. Tuchmann-Duplessis, H.:  Teratogenic Action of Drugs.  Pergammon  Press:
     New York; 1965.
21.Belisle, R. J. and Long, S. Y.:  Tolbutamide treatment  of  pregnant mice:
     repeated administration  reduces fetal lethality. Teratology 13: 65—70,
     1976.
22.Neubert,  D., Merker, H.J., Kohler,  E., Krowke, R. and  Barrach,  H.J.:
     Biochemical aspects of  teratology.  In: Advances in the Biosciences,
     G.Raspe, Ed., Pergammon Press: Oxford; pp. 575-622, 1971.
23. Bakay, B. and Nyhan, W; L.: Effects of Thalidomide and Chlorcyclizine on
     the biosynthesis of nucleic acids and proteins in fetal and maternal tissue
     of therat.J.Pharmacol.Exp.Ther. 171(1): 109-117,1970.
24. Schardein, J. L.: Drugs as Teratogens. CRC Press:  Cleveland; pp.  9—12,
     1976.
25.Kelsey, F. O.: Present guidelines for teratogenicity studies in experimental
     animals. In:  Congenital  Defects,  New  Directions in Research, D.T.
     Janerich, R. G. Skalko and I. H. Porter, Eds., Academic Press: New York;
     pp. 195-204,1974.
26. Golberg, L. M. B.: Discussion pp. 53-55. In: Methods for Detection of
     Environmental Agents that Produce  Congenital Defects, T.H. Shepard,
     J. R. Miller, and M. Marois, Eds., North-Holland Publishing Co.: Amster-
     dam; 1975.
27. Wilson, J. G.: Methods for administering agents and detecting malformations
     in experimental animals. In: Teratology: Principles and Techniques, J. G.
     Wilson  and J.Warkany, Eds., University of Chicago Press: Chicago; pp.
     262-277,1965.
28. Staples, R. E. and Schnell, V. L.:  Refinement in rapid clearing technique in
     the KOH-alizarin-red-S method for fetal bone. Stain Technol. 39: 61—63,
     1963.
29. Hutchings, D.  E.:  Behavioral  teratology:  embryopathic  and  behavioral
     effects  of  drugs during  pregnancy.  In: Studies on the Development of
     Behavior and the Nervous System, Vol. 4, Early Influences, G. Gottlieb,
     Ed., Academic Press: New York; pp. 7—34, 1978.

-------
                                    115

30. Vorhees, C. V., Brunner, R. L. and Butcher, R. E.: Psychotropic drugs as
      behavioral teratogens. Science 205: 1220-1225, 1979.
31.Buelke-Sam, J.  and Kimmel, C. A.:  Development and standardization of
      screening methods for behavioral teratology. Teratology 20: 17-29, 1979.
32. Kimmel, C. A.:  A profile of developmental toxicity. In: Developmental
      Toxicology, C. A. Kimmel  and J. Buelke-Sam, Eds., Raven Press; in press,
      1981.
33. Rice,  J. M.:  Perinatal period and  pregnancy:  intervals  of high risk for
      chemical carcinogens. Environ. Health Perspect. 29: 23-27,1979.
34. Spyker, J. M.: Assessing the impact of low-level chemicals on development:
      behavioral and latent effects. Fed. Proc. Fed. Am. Soc. Exp.  Biol. 34:
      1835-1844,1975.
35.Chernoff,  N. and  Kavlock, R. J.:  A potential  in vivo  screen for the
      determination  of  teratogenic  effects  in mammals.  Teratology  21:
      33A-34A, 1980.
36. Johnson, E. M.: A subvertebrate system for rapid determination of potential
      teratogenic hazards. J. Environ. Pathol. Toxicol. 4: 153—156, 1980.
37. Best, J. B., Morita,  M., R.agin, J. and Best, J., Jr.: Acute toxic responses of
      the freshwater planarian, Dugesia dqrotocephala,  to methyl-mercury. Bull.
      Environ. Contarn. Toxicol.; in press, 1981.
38. Best, J. B., Morita,  M.  and Abbotts, B.:  Acute  toxic responses of the
      freshwater planarian, Dugesia dorotocephala, to chlordane. Bull. Environ.
      Contam. Toxicol.; in press,  1981.
39. Bournias-Vardiabasis, N., Terplitz, R. L. and Seecof, R. L.: An in vitro assay
      for teratogenesis.  Teratology 21: 29A, 1980.
40. Streisinger, G.: Invited discussion on  the possible use of zebra fish for the
      screening of teratogens. In:  Methods for Detection of Environmental
      Agents that Produce Congenital Defects, T. H. Shepard, J.R. Miller and
      M.Marois, Eds., North-Holland Publishing Co.: Amsterdam;  pp. 59—61,
      1975.
41. Wilk, A. L., Greenberg, J. H., Horigan, E. A., Pratt, R. M. and Martin, G. R.:
      Detection of teratogenic compounds using differentiating embryonic cells
      in culture. In Vitro 16: 269-276, 1980.
42. Braun, A. G., Emerson, D. J. and Nichinson, B. B.: Teratogenic drugs inhibit
      tumor cell attachment to lectin-coated surfaces. Nature 282: 507—509,
      1979.
43. Filler, R.:  An in vitro/in vivo  coupled prescreen to  identify teratogens
      requiring metabolic activation. Teratology 21: 37A, 1980.
44. Kochhar, D. M.: The use of in vitro procedures in teratology. Teratology 11:
      273-288, 1975.
45.Neubert, D. and Barrach, H. J.: Significance of in  vitro  techniques for the
      evaluation of embryotoxic  effects. In:  Methods  in Prenatal Toxicology:
      Evaluation of Embryotoxic Effects in Experimental Animals, D. Neubert,
      H. J.  Merker and T. E. Kwasigroch, Eds., Georg Thieme:  Stuttgart; pp.
      202-209,1977.

-------
                                    16
46. Manson, J. M. and Simons, C. R.: In vitro metabolism of cyclophosphamide
     in limb bud culture. Teratology 19: 149-158, 1977.
47.Kochhar, D. M. and Agnish, N. D.: Teratogenic testing in vitro. In: Toxicity
     Testing In Vitro, R. M. Nardone, Ed., Academic Press: New York; in press,
     1981.
48. New, D. A. T.: Techniques for  assessment of teratologic effects: embryo
     culture. Environ. Health Perspect. 18:  105-110, 1976.
49. Brown, N. A., Goulding, E. H. and Fabro, S.: Ethanol embryotoxicity:
     direct effects  on mammalian embryos in vitro. Science  206:  573—575,
     1979.
50. Klein, N. W., Vogler,  M. A., Chatot, C. L. and Pierro, L.J.: The  use of
     cultured rat embryos to  evaluate  the teratogenic activity  of serum:
     cadmium and cyclophosphamide. Teratology 21: 199-208,1980.
51. Johnson, E. M.: Screening for teratogenic potential: are we asking the proper
     question? Teratology 21: 259,1980.
52. Staples, R. L.: Teratogens and the Delaney Clause. Science  185: 813, 1974.
53. Young, J. F.  and Holson, J. F.: Utility of pharmacokinetics in  designing
     toxicological  protocols and   improving  interspecies  extrapolation.  J.
     Environ. Pathol. Toxicol. 2: 169-186,1978.
54.Jusko,  W. J.: Pharmacodynamic principles in  chemical teratology: dose-
     effect relationship. J. Pharmacol. Exp. Ther. 183: 469-480,  1972.

-------
                          CHAPTER 5
                         ••«M^HI^
                OTHER CONSIDERATIONS:

        EPIDEMIOLOGY, PHARMACOKINETICS,

                 AND SEXUAL BEHAVIOR


             Epidemiology: Methods and Limitations

    Epidemiology has been defined as  the study of the distribution
 and determinants of disease and injury in humans.  It focuses on the
 occurrence of disease in groups of individuals or populations rather
 than in any single individual (1). Ideally, reproductive and teratologic
 effects of environmental agents would be assessed  in epidemiologic
 studies of human populations because of the difficulties inherent in
 extrapolating from other  species. However, ethical considerations
 render randomized controlled trials generally unfeasible.  If  one
 cannot  experiment, then  one can only  observe,  but in some
 circumstances even observation is not possible (e.g., risk assessment
 of  new  chemicals  before  they  are introduced into  the  human
 environment).
    Observational epidemiologic studies can be classified into those
 that generate hypotheses and those that formally test hypotheses and
 quantify risks. The following list is not an exhaustive delineation of
 all  possible  epidemiologic approaches in these categories,  but it
 includes those which  may  be most useful for environmental  risk
 assessment.

 Hypothesis—generating studies

    Case reports are a source for raising suspicions about substances
 that might be teratogens or reproductive hazards  (e.g., an  astute
clinician's  association  of thalidomide with phocomelia). However,
only very striking or  very rare outcomes can be  detected  in  this
manner and  often after a considerable  length of time. For the vast
majority  of pregnancy  outcomes,  formal  epidemiologic studies
involving appropriate comparison groups are required.

-------
                                118

    Correlational studies evaluate  the  patterns of  morbidity  or
mortality in populations where classification is made on the basis of
aggregates of individuals as  distinct from single individuals (e.g.,
geographic-specific  spontaneous abortion rates correlated with area-
specific air pollution levels).
    In demographic studies, routinely collected  information is used
to  estimate  disease rates in  populations composed  of individuals
classified by limited demographic characteristics (e.g., age and sex),
allowing for the identification of subgroups at particularly high risk
and of changes in the rates over time.
    Population-based registries can detect changes in the incidence of
the outcomes being registered (e.g.,  spontaneous abortions, low birth
weight, birth defects, neonatal deaths). If placed in selected areas of
suspected high risk and "clean" areas of presumed low risk, they may
point up differences potentially resulting from environmental causes.
Their case materials are a useful resource for mounting case-control
studies of suspected environmental hazards (see below).


Analytic studies for formally testing hypotheses and
       quantifying risks
    In the case-control design, a series of individuals with an observed
effect (the outcome of interest) and a series of unaffected individuals
are compared  with respect  to their previous  exposure  to  the
environmental  agent of interest. The case-control method is most
appropriate  for the study  of  extremely rare  outcomes, such  as
ambiguous genitalia and specific birth  defects, and  relatively  rare
outcomes, such as infertility  and ectopic pregnancy. It is not useful
for the study of very rare exposure  unless the study is conducted in a
 selected  setting with sufficiently large numbers of exposed persons
 (e.g., occupational settings). Case-control studies are not feasible
 when previous exposures cannot be  ascertained.
    In cohort studies, cohorts of exposed and unexposed individuals
 are followed  for  the subsequent  occurrence  of the outcomes  of
 interest. This design may be  the most desirable method for studying
 fairly  common outcomes  (e.g.,  spontaneous abortions),  for deter-
 mining conception rates, and for  evaluating subtle  indications of
 reduced fertility, such as variations  in interpregnancy interval.

 Limitations
     All  observational  studies  are  subject  to  certain limitations.
 Studies  classified  as  hypothesis-generating lack  information  on

-------
                                119

potential distorting factors;  for this  reason,  among others, they
cannot  be  used to  establish  cause-effect  relationships.  Formal
analytic studies do have this capacity if they are valid (i.e., relatively
free  of biases  attributable to  selection, measurement,  and con-
founding).
    If losses to  follow-up are related to outcome status  (cohort
studies) or if  entry into the study is related to  exposure status
(case-control studies), then selection bias will be present. This can
occur in industrial settings,  for example,  if individuals who  are
exposed to hazardous substances tend to leave  the industry and
become lost to follow-up because they become ill.
    Errors  in  measurement of exposure or outcome, if unequal
between the groups being compared, can lead to overestimation or
underestimation  of an effect. Equal measurement errors will always
lead  to  attenuation of an effect, and this is a particular problem
when exposure or outcome is difficult to measure, as is the case with
many environmental exposures and some reproductive outcomes. In
cohort  studies, there is particular concern that the ascertainment of
subsequent outcome be unbiased, while in case-control studies, there
is particular concern that the  measurement of prior exposure to  the
agent of interest be unbiased.
    Because  observational  epidemiologic studies  deal with non-
randomized populations, a central  concern is  whether the groups
being compared  are similar in all relevant  characteristics.  If they
differ in factors related to both the exposure and the outcome, then
confounding bias will be present.  Properly conducted epidemiologic
studies  will make allowance for all known risk factors of the health
outcome of interest, either in the design or in the analysis.
    For  assessment  of the validity  of  a particular study, detailed
information  about  recruitment  and participation  of the study
population,  measurement of the parameters of interest, method of
analysis, and efforts to assess potential biases must be available. A
single epidemiologic study, even if valid, can seldom by itself rule out
chance  or bias  as  the  explanation  of an  observed association.
Establishment of causal associations usually  requires the accumula-
tion of consistent evidence  from valid studies of human populations.
That cause precedes effect must also be  demonstrated. Belief in a
particular hypothesis can be strengthened by evidence from animal
studies,  by  a  biologically  plausible mechanism, and by  a dose-
response relationship.  In general, the stronger an association, the
easier it is to  establish a causal association. Conversely, the  smaller
the effect, the more difficult it is to  demonstrate. Failure  to detect

-------
                               120

an effect may simply reflect inadequate sample size or insufficient
time for the outcome to become manifest.
   This brief  review touches on  only  a few of the  potential
limitations of observational epidemiologic studies. The assessment of
the validity of any particular study requires extensive knowledge of
epidemiologic methods and experience with their application.
Possible data sources and useful approaches
    Currently  few epidemiologic studies  attempt to detect  human
teratogenic  and reproductive hazards or to quantify their effects.
Furthermore,  there  is no  systematic  application of epidemiologic
methods for this purpose. It would be desirable to have programs
specifically designed to raise suspicions and to test hypotheses. To be
effective, these programs must be supported on an ongoing basis.
    Before pilot  testing  any new  epidemiologic program, however,
the potential usefulness of existing studies and data bases should first
be evaluated. Several examples of potentially useful systems are given
below.
    There are  several registries of birth defects in the United States,
for example, the Birth Defects Monitoring Program of the Center of
Disease Control, which collects information from selected hospitals
throughout the country, and the birth defects registries of metro-
politan Atlanta and of Nebraska and Florida.  The  development in
selected regions of population-based registries of reproductive health
outcomes (including birth defects, ectopic pregnancies,  and  sponta-
neous abortions)  could  point to  potential  environmental hazards.
Even in a particular geographic area, clusters of cases or changes in
rates over time could suggest a source of environmental contamina-
tion.
    Vital statistics have been analyzed from time to time, depending
upon the interest of the investigator. For example, infant mortality
rates have correlated with chlorination levels of public water supplies
in  New York (1).  In addition, vital statistics have  been used as
indicators of reduced male fertility in an occupational setting (2, 3).
A systematic  ongoing analysis of vital statistics  data  in relation to
routinely  collected  environmental data might be  quite useful  for
raising suspicions about environmental hazards.
    Two  ongoing surveillance systems  based  on  the  case-control
approach are  currently in operation:  one is designed to discover and
to  evaluate adverse  drug effects that are serious enough to warrant
hospitalization (4, 5), arid the second is directed to the discovery and

-------
                                121

 evaluation of drug teratogenic effects (6). In principle this meth-
 odology is applicable to the discovery of environmental agents that
 are  reproductive  hazards or  teratogens.  For  the surveillance  of
 occupational exposures, programs could be located in specified areas
 of the country where occupational exposures to suspected hazards
 are high. The application  of this methodology to the  study  of
 nonoccupational environmental exposures is more difficult, in part
 because  individuals  may not  be  aware  of what they have been
 exposed to.
    The  cohort method has been  used to identify several health
 hazards in occupational cohorts (7, 8). Other cohorts that  might  be
 useful are enrollees hi health maintenance organizations (HMOs) (see,
 for example, Ref.  9). However, an HMO data base has the limitation
 that only outcomes that come to medical attention can be studied.
 Moreover, no HMO currently has a  computerized data base  in a form
 that would be useful for the  conduct of epidemiologic studies  of
 environmental exposures. A large investment would be required to
 build and maintain such a data base.
                        Pharmacokinetics

    Pharmacokinetics can be defined as the quantitative study of the
absorption,  disposition, metabolism, and  elimination  of drugs,
poisons,  and other chemical  agents in the body. It is important to
evaluate  pharmacokinetic variables at different doses and routes of
exposure to understand the toxicological significance of exposure.
    Pharmacokinetics can be employed  for at least two  purposes:
definition of the  concentration levels of the agent in blood  or in
tissues where the site of action is presumably located, and quantita-
tive description  and prediction of the relevant concentration levels,
usually with a mathematical model. The first should be routinely
done   to  the extent possible as  an  aid  in  interpreting  other
measurements being made.  The second requires much more compre-
hensive study but has the  potential for predictive purposes such as
risk assessment.
    At this time,  there are  several  basic textbooks of pharma-
cokinetics: Notari (10) presents an elementary overview, but with,
many applications; Wagner  (11) and Gibaldi and Perrier (12) provide
collections of the basic mathematical models and solutions with
illustrations of their use. These classical treatments permit organiza-
tion of pharmacokinetic data, along with some biological interpreta-
tions  of  amounts  of an agent in the  "central" regions of the  body

-------
                               122

(blood, vital  organs) versus "peripheral" regions  (other  tissues).
However,  for use of measured levels  in specific  tissues,  models
incorporating what is known about quantitative aspects of anatomy
and physiology have been found useful (13, 14). Another important
feature of this alternative  approach is  to  enable  use  of known
physiological  and  pharmacological  differences between  animal
species to  define  some of  the critical  parameters  for quantitative
extrapolation to man.  A review  is  given  by Dedrick  (15),  and
suggestions  for defining  similarities between animal species  are
described by Dedrick and Bischoff (16).
   A survey of application of the above pharmacokinetic approaches
to some areas of toxicology is given by Gehring  et al. (17), and
further discussions are  in chapters of  World Health Organization
Environmental Health Criteria (18)  and  Filov  et al. (19). Some
specific issues of importance concerning reproductive and teratogenic
effects are described by Young and Holson (20).
   When  pharmacokinetics is applied  to the specific  area of
teratology,  the  major determinants of the teratogenic  agent's
reaching and accumulating in the conceptus are the usual aspects of
pharmacokinetics  in the mother, plus the  unique features of
transplacental transport, and pharmacokinetics in the conceptus. The
maternal pharmacokinetics may be monitored by the blood half-life
(20), although it may also be desirable to have more complete details
of the disposition into the uterine tissue, as well as the presence of
active metabolites and inducible  catabolic enzymes,  later mobiliza-
tion of stored agent, and  any differences between pharmacokinetics
in chronic versus  acute exposures. The uptake and disposition into
the  conceptus may be  partially predicted from  knowledge of
placental membrane transport parameters (models  of oxygen and
glucose  transport  may  be a useful  basis  [22]), specific  and
nonspecific binding, and other features  of the developing fetus. It is
crucial to  determine  these effects  during the period  of major
organogenesis.
    Few available studies  have applied pharmacokinetic principles to
specific areas of female or male reproductive organs,  especially with
reference to formulating  models that could be used for predictive
purposes. In one of the few, uptake  of cancer chemotherapeutic
agents into the human uterus  has been successfully  described  by  a
physiological pharmacokinetic model,  which was  then  used to
formulate  clinical dosage regimens (22). In another, Lee and Dixon
(23)  present the  results  of their innovative study  of the pharma-
cokinetic determinants of uptake into male gonads.

-------
                               123

    Clearly, much more needs to be done before pharmacokinetics
 can be routinely utilized as an adjunct in better defining the basis for
 risk  assessment  of  reproductive  and teratogenic toxic  effects.
 However, information obtained using the reasonably well developed
 methods described in the earlier references should aid in developing
 methods to resolve some of the issues.


                         Sexual Behavior

 Introduction

    Overview.  The behavioral  aspects of reproduction encompass a
 broad spectrum  of activities including courtship  behavior, sexual
 behavior, parental behavior, and  a variety of social activities that
 subtly influence the probability of reproductive success. The scope
 of  this  discussion is limited primarily to sexual behavior  for the
 following reasons.
    • This  behavioral aspect of reproduction has received the most
      detailed and extensive attention from clinicians and laboratory
      investigators.
    • Evaluation procedures  for sexual behavior  of  a variety  of
      animal species are well established.
    • Choices can  be  made  among several  existing standardized
      procedures currently in use in laboratories around the country.
    • Observational methods are simple and direct,  and workers at a
      moderate level of skill can be quickly trained to obtain reliable
      measurements.
    • Many of the testing procedures recommended in the following
      presentation  can  provide  insight  into the  probable  locus  of
      action of the putative toxicant, and this could not be as easily
      achieved if the scope of the  investigations was extended  to
      include at this time other behavioral aspects of reproduction.
    The presentation that follows attempts to establish the impact
and causes of sexual dysfunction in humans, to discuss methods of
assessing human  sexual behavior,  and to indicate the  difficulties
associated  with  investigations of human sexuality when there are
neither  controls  nor  standard norms.  The  evaluation of sexual
behavior patterns in animals are presented as simple tallies of specific
motor responses; however, it will be emphasized that many elements
of human sexual behavior are unique, having no animal counterpart,
thus making  uncertain  any extrapolation of data on animal sexual
behavior to humans.

-------
                               124

    Definition and scope.  The study of sexual behavior encompasses
the measurement of normal and abnormal function as well as the
identification  of the factor(s)  responsible for  the  impairment of
sexual behavior. The quantitative measurement of sexual functioning
involves the establishment of norms or averages for groups and for
the individual and most often focuses on coitus itself. For humans
more extensive and varied measures are necessarily employed, which
include sexual imagery,  sexual fantasy, varieties  of overt sexual
experience, self concept  and gender  identity, assessment of inter-
personal relationships, choice of sexual  object,  and level of sexual
skill.
    For the most part, animal models available today do not provide
data that might be required for assessing human sexual functioning
and for identifying the factors responsible for imparied expression of
sexual behavior. Nevertheless, important  advances made in the study
of animal sexual behavior provide at least for the initial screening of
toxicants that could deleteriously affect human sexual conduct. To
identify  the  factors  responsible  for  normal and  impaired sexual
behavior, investigators  of  animal sexual behavior  have identified
three  separate components: sexual  attractiveness, sexual initiative,
and sexual responsiveness. The current working assumption is that
these factors have a much broader generalizability across species than
any isolated  species-typical behavioral  response or activity (e.g.,
mounting). The evidence  and  arguments  favoring this  working
assumption have been set forth persuasively by Beach (24), who uses
the alternative terminology  of attractivity, proceptivity, and  recep-
tivity  to  designate the  three factors. Accepted  systems of measure-
ment  have been worked  out for a variety of laboratory animals
including the rat and macaque monkeys (25—27).
    It should be realized from  the outset that  manifest  sexual
behavior reflects the functional integrity of a broad system com-
prising elements of drive and reward, perception, sensory function,
motor performance, the physicochemical actions  of gonadal hor-
mones on neural and somatic tissues, and,  finally, central nervous
system processing and coordinating of the interactions of all of these
elements. An efficient  screening system utilized for detection of
toxic effects should attempt at some stage to distinguish between
general motoric disability (ataxia) and impairment of specific sexual
reflexes; between general lassitude and loss of specific sexual interest
 or motivation; and  between  the impaired  production of gonadal
 hormones and the impaired actions of these hormones. At this tune,
 however,  no simple and  efficient test  of  sexual behavior or tests

-------
                                125

 designed  to  measure  sexual  attractiveness, initiative, and respon-
 siveness  automatically  determine  whether elements  of the  broad
 system are impaired  either as a result  of general debilitation or
 selectively and specifically "with  regard to  sexual  performance.
 Currently  available tests, while not permitting decisions about the
 specificity of the effect of a toxic substance,  can serve  as  early
 warning signals that normal reproductive function has been impaired.
    Impact of sexual dysfunction on humans.  Data on the incidence
 of  human sexual dysfunction  and  its spontaneous  remission are
 neither extensive  nor  very  reliable.  Certainly the most common
 clinical problems are primary and  secondary  anorgasmia in females
 and ejaculatio praecox  and erectile  impotence in  males.  These
 disorders,  however, represent  extremes of  dysfunction that are
 unacceptable to most humans, and those so affected commonly seek
 clinical help.  Less extreme forms of inadequate sexual response
 clearly exist and  are  often  tolerated, but only in the  sense that
 professional counseling is not sought. Even  though many individuals
 are reluctant  to  seek  professional help,  the  importance of sexual
 behavior and  sexual gratification to the overall quality of life and
 individual  well-being  is  generally recognized. Many individuals are
 willing to relinquish reproductive capabilities (through vasectomy or
 other contraceptive  means)  to  enjoy fewer restrictions on sexual
 activity. Few people, however, will relinquish  sexual gratification to
 gain contraception.
    The importance attached to  sexual gratification by  individuals in
our society implies that sexual inadequacy, even when tolerated, may
not be without serious consequences. Our monogamous social system
depends in a very fundamental way upon a  sexual contract between
two individuals. Failure to achieve, or even reduction of, sexual
satisfaction seriously threatens the interpersonal relationship, as the
growing number of marriage counselors recognize. How much of the
growing sexual and marital discord is attributable to  sociopsycho-
logical factors and how much might be attributable  to  disturbances
in the physiological systems underlying sexual performance is not
known. The possibility exists, however, for toxic substances in the
environment  to  cause  disturbances  in  sexual performance and
thereby contribute to interpersonal discord.
    Causes of  impaired sexual performance.  Sexual dysfunction in
human  beings is  poorly understood.  Many  sexual disorders are
primarily psychogenic  and respond  well to psychological treatment.
Others that  are  resistant to  psychological  approaches  seem  to
originate in specific genetic factors, early  experience, or a combina-
tion of genetic and experiential determinants.

-------
                               126

   Endocrinopathies, especially abnormalities of the gonadal hor-
mones,  have marked influences on the  pattern of sexual behavior
displayed by  animals,  and  although their  influence  is less well
described for humans, it cannot be said  that their role is negligible.
For  both the male  and the female, inadequate gonadal hormone
activity  commonly  results  in  deficient sexual performance.  Sub-
normal   effectiveness of  gonadal  hormones can  be  due  to (a)
deficiencies in production, (b) deficiencies in bioavailability, and (c)
deficiencies in target organ sensitivity and/or responsiveness.
   Androgen deficiency affects male behavior in two distinct ways.
First, during early stages of development  (probably before birth in
humans) deficiencies in androgen lead to incomplete development of
central  neural  and  peripheral  somatic structures essential for the
expression  of  masculinity and male behaviors including,  but not
limited  to,  male sexual behavior.  Second, during adolescence and
adulthood,  deficiencies  in  androgen  are  associated with  reduced
sexual responsiveness and sexual initiative.
    Behavioral disorders associated  with  excessive androgen have not
been identified for the male, although there are recurrent suggestions
that excessive amounts during early  stages of development lead to
permanent  androgen insensitivity. In  the female, however, excessive
androgen during early developmental stages leads to the development
of masculine behavioral and somatic characteristics and,  in  some
species,  to  the suppression or  loss of feminine  behavior traits. This
suppression  of feminine traits can  include  sexual  behavior,  and
female  sexual  responsiveness can  be only reduced.  In adulthood,
excessive androgen in  female humans may  lead  to  measurable
somatic virilization  (such as hirsutism  and  clitoromegaly) without
any marked effect on psychological and behavioral traits. Increases in
sexual  initiative  and  responsiveness may  be large  enough to be
distressing and disruptive to an established interpersonal relationship.
In nonhuman primates, androgens have also been implicated in the
control  of  proceptivity (sexual initiative), but not in the control of
receptivity.  In other mammalian animals, excessive  androgen in
adulthood may cause a sharp  increase in  the frequency of malelike
mounting activity  and  aggression  with  or without  concomitant
alterations in female sexual behavior. Most of the psychological and
somatic changes induced  by excessive  androgen in adulthood  are
partially or totally reversible when the hormonal excess is eliminated;
however, some of  the more dramatic  somatic changes (e.g., voice
 changes and hirsutism) are irreversible.
    Estrogen  and  progestagens play   essential but incompletely
understood roles in the regulation of female sexual response. These

-------
                                127

hormones are produced and secreted in much larger amounts by the
ovaries than by the testes or the adrenals in normal  physiological
conditions. Whereas human female sexual behavior does not depend
entirely  upon the actions of estrogens and progestagens, its expres-
sion  is greatly  facilitated  by  their actions on  both  central and
peripheral  neural and  somatic  tissues.  In  animals, especially  the
common laboratory  forms, the ovarian  hormones are much more
essential to the  expression of female sexual behavior than in human
beings. Generally, the effective estrogen is estradiol and the effective
progestagen  is  progesterone.  These  two  steroid  hormones  act
synergistically in the induction  of both proceptivity and receptivity
in rats, mice, hamsters, and guinea pigs. The two hormones may also
act  antagonistically,  however,  and which  relationship obtains  de-
pends upon whether or not the estrogen has been free to act for a
specifiable period  of time  without any concurrent  actions of  a
progestagen. The synergistic relationship depends upon the sequen-
tial action of an estrogen followed (usually 36 to 48 hours later) by
the  action of a progestagen. An antagonistic  relationship will be
evidenced whenever a progestagen and  an estrogen are  both present
throughout the period of observation or study.
    Excessive estrogenization acts to lengthen the period or duration
of receptivity and proceptivity. An established  norm of eight hours
for the duration of receptivity in a colony of rats can be extended to
12  or 14  hours  by excessive  estrogenization. In extreme cases,
excessive estrogenization can extend receptivity indefinitely.
    Excessive progesterone has no measurable effect if the period of
stimulation  is  brief. If the  period  of excessive  stimulation  is
protracted, however,  receptivity and proceptivity can be indefinitely
suppressed or inhibited. The antagonistic effect of progestagens is
transitory and reversible when these hormones  are brought back to
physiologic concentrations.
    It  is difficult but  not impossible to distinguish between the
antagonistic effects of excessive progestagen and  a deficiency in
estrogenization.   A  deficiency   of  estrogen,  like  excessive  pro-
gestagenization,  has the primary characteristic of weak or absent
female sexual response. A  distinction  between  the two possible
causes of impaired sexual response can be made by institution of
appropriate experimental hormone administration to ovariectomized
females.
    Excessive  estrogen  or  progestagen  during early  periods  of
development  can produce permanent deficiencies in female sexual
response  in  a variety of laboratory animals. Comparable  data for
human beings do not exist.

-------
                               128
    Sequelae of estrogen and/or progestagen excess or deficiency in
males have not been well worked out. Supraphysiologic levels of
estrogen have been administered to human males in cases of prostatic
cancer. Sexual drive and erectile potency sometimes decline in these
cases, presumably because the  estrogens block the release by  the
pituitary of testis-stimulating hormone, and an androgen deficiency
results. Similar effects could be  obtained in some laboratory animals
(the guinea pig), but  not in  others  (the rat,  in  which excessive
estrogens  mimic androgens  in the  potentiation  of male  sexual
activity).
    The effects of hormone excess and deficiency could occur when
chemical substances mimic or antagonize physiological actions of the
relevant gonadal hormone. Other chemical agents could enhance the
degradation of steroidal  hormones in the liver or kidneys and thereby
reduce or limit their effectiveness. Still other chemicals could either
act on the hypothalamic-pituitary system to modify the release of
trophic substances essential for the normal production of the gonadal
hormones or act directly upon  the glandular tissues responsible for
their production.
    Many  factors, aside  from  alteration of  or interference with
hormonal support, can  act to impair sexual behavior. These factors
are difficult to assess in standard laboratory tests, either because no
suitable animal model can be found  or because appropriate assess-
ment  would  involve  procedures  too  elaborate  and  costly  for
routinization. Although testing for alteration or interference with the
hormonal support of sexual behavior assesses only a limited aspect of
requirements for human sexual adequacy, it has the advantage that
the information gained  is reliable,  quantitative, and amenable to use
in estimating the risk to human sexuality posed by specific chemical
substances.

Qualitative evaluation of risk potential
    Interspecies comparisons. Requirements for genetic variability in
the test model animal  that  approximates that  encountered in the
human  population have already been discussed in the section titled
"Interspecies Comparisons" in Chapter 4. Highly inbred strains ought
to be generally avoided  unless several are used to determine the range
of sensitivity to the test substance.
    Known and suspected differences among species in the manner in
which gonadal hormones regulate sexual behavior mandate the use of
more than one species. For example, the major androgen produced
by the testis is  testosterone in most  mammals.  This  hormone is

-------
129
           the  normal  male rat,
           or  via  conversion to
           mediating male  sexual
 metabolized within somatic  and neural cells  to  a variety of other
 steroids including estradiol and dihydrotestosterone. In some species,
 like the rat, the estradiol derived from bioconversion of testosterone
 is a potent stimulator of male  sexual behavior in  the adult  and a
 potent masculinizer of the developing brain in the fetus and neonate.
 In contrast, in the guinea pig this estrogen metabolite of testosterone
 is without any measurable stimulating effect on male sexual behavior
 when it is given to castrated adults. The view is widely held that the
 display of male  sexual  behavior depends upon the intracellular
 conversion of testosterone  to   estradiol in
 whereas  testosterone  acts  either  directly
 dihydrotestosterone  on the  neural  tissues
 behavior in the guinea pig.
    The "rat  model" for cellular  utilization  of testosterone  (by
 conversion to an estrogen) is  valid for hamsters and some but not all
 inbred strains of mice. The "guinea pig model" is valid, based on very
 limited data, for the rhesus monkey and also for humans.
    This species  difference is important because erroneous conclu-
 sions are possible if testing is limited to a single species. Any putative
 toxicant that blocks  intracellular aromatizing enzymes needed  for
 bioconversion of  testosterone to estrogen  would impair adult male
 rat sexual behavior, but the same compound would not likely have
 an effect  on sexual behavior of male guinea pigs, rhesus monkeys, or
 humans.
    Other species  differences, too numerous to detail here, include
 differences in the role of specific neural structures, in the contribu-
 tion of specific neurotransmitters, in the amount and kind of carrier
 protein that is present in the bloodstream and binds and transports
 the  steroid hormones, in the chemical structure of the pituitary
 trophic hormones, and certainly in the form and normal frequency
 of sexual expression. All of these  species differences argue for the use
 of more than  one species in screening for toxicity as well as  for
judicious choices when only a  few are to be used. In short, the choice
 of a species to use  as a model animal places  profound and subtle
 limits on  evaluating the toxic consequences of any chemical agent,
 and these limits have to be reckoned with.
    Certain relatively simple and  easy-to-conduct tests could serve as
 a preliminary screen to indicate the degree of likelihood of an agent's
 affecting either the early sexual differentiation or adult expression of
 sexual behavior. Based on the assumption that chemical  substances
 that pass  the  placenta and gain  access to the fetal tissues are more
 likely  to  affect  early  development  than  those  that  fail to pass

-------
                               130

through the placenta, a relatively simple and efficient study of the
distribution of the radioactively labeled chemical substance could be
carried out. It is also reasonable to use radiolabeled material in adult
animals to determine whether the substance crosses the blood-brain
barrier and has the potential of acting directly on  nervous tissue.
These simple tests,  of course, are not specific indicators that sexual
behavior would be altered by the putative toxicant. Positive findings
from  these tests would  merely serve the purpose of  indicating
increased likelihood.
    Other considerations. It is reasonable to  assume that a wide
variety of other factors are important in facilitating extrapolation of
animal tests of a toxicant to the estimation of risk to humans. These
include dosages of  putative toxicant  used, route of administration,
duration and frequency of exposure, species thresholds and sensitivi-
ties to  the chemical substance, and  specific pharmacokinetics and
pharmacodynamics  of  the  test compound. Whenever information
exists for humans on any of these factors, either for the specific test
compound or a closely related substance, an effort should be made
to select an animal model that most closely parallels the human to
increase the applicability of the animal test results.
    Despite the advantages of objectivity,  ease of  administration,
reliability, and wealth  of background information,  tests of animal
sexual behavior in the present context have severe limitations. First
and foremost  is the high degree of uncertainty  that results of animal
tests could be extrapolated to human sexual behavior. It is likely that
extrapolation  would be  good if a  putative  toxicant  completely
blocked the neurological actions of the sex hormones (especially the
androgens), since hormonal support for sexual  behavior and for the
fetal differentiation of sexual and/or  sex-related behavior is a factor
common to both animals and humans. However, humans and animals
differ  greatly in the numbers and  kinds  of nonhormonal  factors
influencing the  expression of sexual  behavior.  Accordingly, when a
putative toxicant  acts only  on  one or a subset of nonhormonal
factors, there is a strong likelihood that animal test results will not
correspond to effects (or lack thereof) on human  sexual expression.
    A second  area of concern is the nearly total lack of background
information on effects of  known toxicants on sexual behavior in
either animals  or  humans.  This deficiency   thwarts any present
attempt at formulation of procedures for quantitative risk assessment
based on findings from animal tests. This situation can be remedied
only  by providing encouragement of the appropriate research on
animal  models as well as intensive studies of humans  exposed to
known toxic agents.

-------
                                131
Animal studies
   Evaluation  of sexual behavior in adulthood.  Observations  of
sexual behavior in adult animals  can be made by easily trained
nonexperts. Useful assessments of the status of sexual behavior can
be made  from  simple  tallies  of the frequency of occurrence  of
specific motor responses and the latent period from the beginning of
a standardized test to the occurrence  of the specific response. These
are  the  operational  measures of initiation,  attractiveness,  and
responsiveness.
   The procedures described in this  section are designed to permit
reliable,  sensitive  analyses  of the  effects of  potentially active
chemical substances on male sexual behavior. A considerable body of
knowledge gathered in  the  last 60  years  reveal's that  the sexual
patterns of rats and guinea pigs can provide such data. Further, the
existence of extensive data  bases on  these two species provides the
possibility  of  a preliminary indication  of mechanism  of action
underlying observed treatment effects, since determinants of various
aspects of these complex patterns have received much study. Should
more extensive  and expensive  testing of a chemical  be indicated,
dogs, nonhuman  primates,  or  other species may be appropriate.
Methods described below can  be adapted to such  species  using
behavioral  testing procedures described by Dewsbury (28) and in the
references therein.
   In all  tests,  one sex should be treated so that treatment effects
may be detected uncomplicated by effects  of the agent's acting on
the opposite sex. Where the probability of a treatment effect is quite
low, it may be more economical to combine procedures for male and
female treatments into a single protocol. However, the risk that three
rather than two studies may  be required if such procedures are used
should be recognized.
   Assessing sexual behavioral  patterns of males. Effects of various
toxicants  can  be  evaluated as they  alter  the  normal complex
behavioral  patterns of  male rats and guinea pigs. There  are some
considerable  advantages to  toxicological inquiry  in  studying the
behavior of  male animals   that  have been  castrated  and  given
physiological hormone replacement and exogenous testosterone. This
procedure obviates the possibility that impaired sexual performance
might be due to toxic insult  to the hypothalamic-pituitary-testicular
axis  or to the  testis itself.  However, inasmuch as castration and
replacement  therapy  are  complicated techniques  in themselves,
testing the intact animal should serve as an adequate preliminary
screen.

-------
                                132
    In studying the  normal copulatory behavior of laboratory rats,
three  classes  of  events  are  generally  distinguished — mounts,
intromissions, and ejaculations. With the first the male mounts the
female  from behind, displays  shallow pelvic thrusting, but neither
gains vaginal penetration nor  displays the stereotyped pattern of
dismounting. Intromissions begin  similarly, but the male achieves a
single  deep thrust and dismounts in a vigorous and stereotypical
pattern. Ejaculations occur only after several intromissions and are
characterized by an  intravaginal thrust that is longer and deeper than
that of intromissions. Sperm  are transferred only on ejaculations.
The male  mounts  the female during mounts,  intromissions,  and
ejaculations,  but the three classes  of events are distinguished as just
indicated.  During  pair mating copulatory events occur in "ejacu-
latory series,"  with  each series terminated  by an  ejaculation and
separated  from a resumption of copulation by  a postejaculatory
refractory period. In standard testing cages, males normally display a
mean of approximately seven  ejaculatory series before attaining an
arbitrary,  but  standard,  satiety criterion of  30  minutes with  no
intromissions or ejaculations.
    Standard measures of male copulatory behavior include mount
latency (ML),  time from  start of a  test to  the first  mount or
intromission; intromission latency  (IL), time from the start of a test
to the first intromission; ejaculation latency (EL), time from the first
intromission  of a series  to its terminal ejaculation; intromission
frequency (IF), number of intromissions in a series; mount frequency
(MF), number of mounts in a series; mean interintromission interval
(Mill), mean interval separating the intromissions within a series; and
postejaculatory  interval (PEI), time from ejaculation to  the  next
intromission. Male receptivity may be quantified by dividing the
number of male chase and follow-bouts by the total  number of
female approaches (29).
    Because various  of these measures can be affected selectively,
specifically, and in  combination,  an accurate interpretation  of a
treatment effect requires a full complement of these measures. As an
example, suppose a  treatment interfered with the process of penile
erection. Males with such  problems often mount females at  rates
much  higher than  normal as they repeatedly  attempt  to effect
intromission. Without a full complement  of measures, such an effect
might  be mistaken for an increase in libido rather than as a deficit.
Similarly, a treatment that alters Mill may secondarily affect IF. In
addition, with  a full set of measures, one can evaluate the control
group  in relation to  animals used in previous studies (see references

-------
                                133

 below) to ensure that it is  providing an appropriate  baseline for
 comparison.
    Full descriptions  of  copulatory behavior in male  rats can  be
 obtained in Beach and Jordan (30), Dewsbury  (27), Larsson (31),
 and Sachs and Barfield (32).
    Copulatory behavior in male guinea pigs differs from that in male
 rats in several important respects. First, whereas rats display but a
 single intravaginal thrust on  each mount with intromission, guinea
 pigs display repetitive thrusts on a single insertion. Second, whereas
 male  rats  rarely,  if  ever,  ejaculate  on  the first  mount with
 intromission,  such  occurrences are more frequent in  guinea pigs.
 Third, although male rats normally display several ejaculations per
 .test  session,  the   occurrence  of  the  first  ejaculation  generally
 effectively terminates copulatory activity in  guinea  pigs. In other
 respects, the copulatory patterns of male guinea pigs are  quite similar
 to those of male rats. Similar measures can be used.
    Descriptions of  copulatory behavior in guinea pigs can be found
 in Young (33)  and Young and Grunt (34).  Various  measures  of
 preliminary aspects of courtship and mating described in  these papers
 may be useful.
    Test of copulatory behavior should be conducted during the dark
 phase of the diurnal cycle. By testing during the second half of the
 dark phase,  behavior generally is more reliable, quicker, and less
 variable — making for a more efficient and sensitive test (35). Tests
 should be conducted at approximately the same time on  each day.
    Males and females should be familiar with the  testing arenas via
 introduction several times on days before test days.
    In tests for male behavior, males are generally  placed  in  the
 arenas for five to ten minutes, after which the female is introduced,
 effectively beginning the test. Tests may be terminated and scored as
 negative if  there is no copulatory activity within a predetermined
 time (e.g., 15 minutes).
   Tests of guinea  pigs should be terminated at the occurrence of
ejaculation.  Those  of rats  should be continued for  two  or three
ejaculatory series. Such tests may require an average of 45 minutes in
rats. It may be feasible to test several pairs of rats simultaneously in
cages close to each other, if the only behavioral patterns to be scored
are those discussed above.
   For reasons of reliability  and  predictability, it is recommended
that  female  mating  partners be brought into behavioral estrus with
exogenous hormones.  Female guinea pigs must first  be ovariecto-
mized; this may or  may not  be done with female rats. For either

-------
                               134

intact female rats or spayed guinea pigs, good results can be obtained
with an intramuscular injection of 0.1 mg of estradiol benzoate three
days before testing and  1  mg  of progesterone approximately six
hours  before  testing.  Somewhat  lower doses can  also  be used.
Females should be placed briefly with a vigorous, nonexperimental
"indicator" male  immediately  before testing  to ensure that the
injection regimen has been effective in inducing receptivity. A single
female rat in estrus can be used to  evaluate sexual performance of at
least three males. A single female guinea pig should not  be used for
more than two males.
    There  are  many factors, both quite  specific and highly non-
specific,  that' can  alter copulatory  behavior. If  there  are gross
increases or decreases in body  weight or  activity levels, changes in
sexual behavior are probably secondary to more general effects. If
body  weight  and  general  activity  is near normal and copulatory
behavior is altered, however, greater specificity of action probably is
indicated. Some indication of the nature and degree of effect can be
determined by considering the constellation of measures altered, the
magnitude of effect, and reversibility. By comparing these changes to
those described in the literature as resulting from other treatment,
some preliminary indication as  to probable mechanism of action can
be gained. Any   alteration requires  some further analysis. Such
subsequent studies may be directed at analyzing neural, endocrine,
and other systems to determine  whether or not the effect seems
appreciable and likely to affect humans.
    Assessing sexual behavior patterns of females. A substantial and
useful background of behavioral data exists for both rats and guinea
 pigs  from a  number of  inbred strains  as well  as genetically
 heterogenous stocks. It is possible to evaluate proceptivity, recep-
 tivity, and attractiveness in female rats in  a single test paradigm with
 a stud male and to evaluate receptivity in the female guinea pig.
    The intact and normally functioning female rat displays a period
 of estrus ("heat") that lasts from  6 to  11 hours  about  every 4 to 5
 days. As long  as  the female is not mated, estrus recurs regularly.
 Recurrent estrus also  occurs in the unmated female guinea pig, but
 the interval between receptive  episodes lasts 14 to 17 days.  In both
 species, sexual behavior depends upon appropriate ovarian secretion
 of estradiol and progesterone. When the ovaries are removed, sexual
 behavior  is no longer displayed. Preceptive and  receptive behaviors
 are displayed in close  temporal proximity and have similar hormonal
 requirements (36).

-------
                               135

    The behavioral response indicative of normal receptivity is the
lordosis posture assumed by the female during mounting by a male
partner. Degree  of receptivity is estimated in a quantitative fashion
by dividing the number of lordosis responses displayed by the female
by the number  of times she is mounted  by her male partner in a
standardized  test. This derived  measure is called  the receptivity
quotient or, alternatively, the lordosis quotient. The measure is more
useful  in  the rat than in  the  guinea pig, because male rats are
normally multiple mounters, whereas male guinea pigs often mount
only once during a mating test. For the female guinea pig, therefore,
an alternative procedure for quantifying receptivity is often used.
The  procedure,  described  fully elsewhere (37, 38), involves manual
stimulation  of the animal's rump  and  perineum by  the human
observer and  measurement of the degree  or duration of the lordosis
response to such stimulation. During mating with a stud male, female
attractiveness may be quantified  by measuring the latency between
introduction of the female and a  male approach, follow, and mount
(29).
   Proceptivity  can be measured  quantitatively in the female rat by
recording the frequency and timing of displays of a variety of motor
patterns including female  solicitations and approaches to the male
partner,  darting,  hopping,  and  ear  vibration.  These preceptive
patterns generally are displayed just prior to the occurrence of male
mounting responses, but they may occur at any time when the male
is quiet or inactive.
    Full descriptions of female rodent sexual behavior can be found
in  Diakow  and Dewsbury  (39);  McClintock  and  Adler  (25);
McClintock, Ansiko, and Adler (40);  and Madlafousek and Hlinak
(26).
   Evaluation  of sexual  responses in the intact female requires
constant and  frequent monitoring  of individual animals. This  is
essential because the occurrence  of the  behavior is restricted to a
short and specific period of the ovarian cycle. The behavior normally
is expressed only during the time the follicle is undergoing its final
preovulatory  swelling.  The ovarian  cycle is  usually  monitored  by
taking  daily vaginal smears for  cytological evaluation, and sexual
behavior usually is displayed  during the proestrous smear or the
transition between proestrous and estrous smears.
   The procedure of monitoring the ovarian cycle by daily vaginal
smears is cumbersome, time-consuming,  and not very precise with
respect to the assessment of sexual behavior. When individual females
are  tested  for receptivity  and proceptivity at an arbitrary time

-------
                               136

relative to  a particular  vaginal  cytology, some  may  be at the
beginning of the period of receptivity, some in the middle, and some
near the end. Others may not yet have reached the receptive stage,
and  for those in various segments  of the  period the  quality of
receptive  behavior will vary accordingly. Furthermore, in the intact
female, impairment  or absence of sexual response  could be due to
impairment of pituitary gonadotrophic activity,  disordered ovarian
production  of steroids,  or impairment of the response  of relevant
neural centers to the gonadal hormones.
    Undesirable variability as well  as uncertainty  about the cause of
impaired  sexual response can be reduced by assessing sexual behavior
in spayed females suitably  treated with injections  of estradiol and
progesterone. Usually  females are  brought  into  good  states of
receptivity by a single subcutaneous  injection of estradiol benzoate
followed  48 hours later with an injection  of progesterone.  All
animals to be tested can then be evaluated at an exact time (usually
six hours) after the progesterone injection.
    The artificial induction of sexual responses has to be done with
precision and with  concern for hormonal stimulation that closely
approximates the normal physiological pattern. Administration  of
excessive amounts  of  estrogen  and progesterone could mask  or
override subtle derangements induced by a toxic substance. If spayed
animals are  used for assessment of sexual behavior, great care must
be exercised to ensure  that  physiological  doses  of estrogen and
progestagen  are administered. Reference to the literature on experi-
mental analysis of female rodent sexual behavior will not be helpful
as a guide to proper hormone treatment, since suprathreshold dosage
regimens are usually used, and these are not appropriate for screening
toxicants. In any  attempt to identify damaging actions of a putative
 toxicant, the investigator should be cautious about  exceeding 1 fig of
 estradiol benzoate and 0.1  mg of progesterone per  adult animal. The
 best general rule to follow is to conduct an initial  parametric study
 on  the specific breed or strain to  be used and  to determine the
 minimum hormonal requirements for induction of estrous behaviors
 in a specified percent of the population.

 Assessment  of human sexual behavior: surveillance and
      epidemiological studies
     Preliminary comments. Direct assessment of human behavior is
 essential  for  evaluating  the  behavioral  effects  of environmental
 toxicants. The extrapolation of animal studies to human behavior is
 limited for  a variety of reasons, (a) Many  aspects of human sexuality

-------
                                137

 and reproductive  behavior are unique and have  no obvious animal
 counterpart (41, 42). (b) While compounds such as steroids do affect
 sexual  motivation in both  animals  and humans, their behavioral
 manifestations  in  humans  are  often  quite different  from their
 manifestations in animals, (c) Human behavior may be disrupted at
 lower toxicant  levels than would be expected from animal studies.
 (d) The exposure of the general population,  but  especially workers,
 to the  compound may be greater in fact than originally estimated
 (also see "Other Considerations").
    We present several different methods for assessing the effect of a
 toxicant on human sexual behavior. Because  direct controls may not
 be possible for practical or ethical reasons, each method has its own
 weakness.  Therefore, we have proposed a variety  of methods and
 suggest that  they be used concurrently if at all possible. This extra
 effort would be justified particularly when the potential benefits of a
 compound are high, but also when animal toxicological screening or
 analysis of the compound's structure indicates that the potential risk
 to human behavior may also be high.  In any case, the behavioral
 assessment procedures for humans need not be cumbersome and can
 be incorporated in any  procedure or  physical  exam designed  to
 monitor the effects of a putative toxicant on reproductive function.
    Behavioral surveillance of humans  potentially exposed to a
 reproductive toxicant. Ideally, new compounds  would be released
 and used  at first  on  a  limited  basis. Then,  changes in  sexual
 satisfaction  and function could be  assessed prospectively with
 adequate  controls. The sexual experience  of the exposed  group,
 perhaps production  workers who  would  be exposed to  higher
 concentrations,  could be compared with a matched group of similar
 workers in an area or plant where the compound  was not yet in use.
 This comparison should be made between two groups of workers in
 the same  plant  or location. If this is not possible, the two  groups
 should  be matched for factors  known to  correlate with  sexual
 attitudes and behavior such  as socioeconomic status, cohort, eth-
 nicity, religion,  and environmental factors. (Industry should  use an
 epidemiologic consultant to determine the matching criteria, sample
 size, and  duration  of surveillance  appropriate for the amount of
 natural variance in the proposed measures of sexual behavior.)
    If limited  release is not warranted ethically or practically and a
general  release occurs,  it  would  be  necessary to  monitor  sexual
satisfaction and the incidence of dysfunction  before as well as after
the compound is released. The large population variability in normal
sexual behavior  may make this  procedure  more sensitive than  a

-------
                               138

comparison between groups. Furthermore, as the behavior of each
person is compared to his own normal pattern, it may be possible to
identify a subpopulation of particularly sensitive individuals.
   These control procedures are essential for evaluating the effect of
a toxicant  on human sexual behavior, because standard norms are
not currently available as a basis for comparison as they are for such
physical variables as sperm  count or menstrual cycle length. As the
number of controlled studies increases, it may be possible to use the
data from  control groups to develop normative statistics for future
evaluations.
   The frequency of sexual intercourse is  not  a good indicator of
sexual satisfaction by itself; it is also necessary to evaluate sexual
arousal, initiation,  and changes in  erotic  imagery and to identify
specific sources of sexual dysfunction. For example, there was little
agreement  about the nature of changes in women's sexual motivation
over the menstrual  cycle until studies focused  on behavior of the
woman herself and her sexual initiation and fantasy rather than on
the frequency of intercourse (43, 44).
    Either  an interview or a short questionnaire can assess sexual
satisfaction and function. It is important  that  the interviewer be
trained in  interview techniques for sexual counseling. Short courses
are  currently  available  for medical  and  lay  personnel in most
academic  medical  centers  (Marriage  Counseling Center  of  the
University   of  Pennsylvania has  a list). Alternatively, there are  a
variety of short  questionnaires  that  correlate  well with  such
physiological  measures of sexual arousability as penile tumescence
and vaginal lubrication (45, 46) and that generate a similar profile
whether completion of the questionnaire is mandatory  or voluntary
(47, 48).
    Another  approach to  the assessment  problem  is based on
epidemiological  data.  The incidence of  cases involving  sexual
dysfunction reported to  such institutions as mental health  clinics,
local physicians, or plant infirmaries can be recorded and used as a
normative   data base. This baseline could be  compared with the
frequency   of reported  cases following  the   release of  a   new
compound. Again,  an epidemiologic consultant should determine
whether the sample from available institutions would be large enough
to detect a toxic effect.
    Evaluation of human  sexual behavior  following exposure  to a
known toxicant. Many compounds have been established as physio-
logical toxicants but have not yet been assessed for behavioral effects
in humans. Estrogenic compounds  such as DBS  and DDT may affect

-------
                                139

 the sexuality of women, while organopesticides that are neurotoxins,
 such as carbaryl, an acetylcholine esterase inhibitor, may affect male
 erectile function.
    If  a population  has been exposed  to such compounds or is
 suspected of being at reproductive risk, behavioral assessment can be
 made at the time that a physiological assessment is being made. The
 same personnel could do this, provided that they have been trained
 in interview techniques. Behavioral assessment under these ex post
facto  conditions  is  particularly  difficult because  knowledge of
 exposure to a toxicant  can distort the retrospective  assessment of
 sexual satisfaction and  behavior.  Therefore, trained  personnel, an
 evaluation immediately  after  the  exposure, and established  norms
 would each help to reduce this bias. In  any event,  an unexposed
 control population should be evaluated using the identical retrospec-
 tive  procedures  and matched  to  the target population for such
 variables as socioeconomic status,  ethnic group, and  local environ-
 ment.
   Risk assessment. If any significant alterations are found, expo-
 sure  to the  toxicant should be discontinued to assess the reversibility
 of the effects. Furthermore, the mechanism of action will need to be
identified to evaluate a risk/benefit ratio. For example, it is possible
 that  erectile function could be impaired through a direct impairment
of cholinergic mechanisms  or indirectly  through  an  increase in
 depression or sense of fatigue (49). Nocturnal  penile  tumescence
 would aid in a differential diagnosis, as erectile function during sleep
is not  impaired by psychogenic factors. Human sexuality is particu-
larly sensitive  to  disruption by many environmental and psycho-
logical factors that are not  specifically  sexual  themselves; most
instances of sexual dysfunction encountered in the clinic are not the
result of a  direct organic cause. Therefore, the mechanisms of any
impairment  of sexual performance or satisfaction will have to be
determined  before a risk/benefit ratio can be assessed.

Priority areas for future research

   Few experimental studies have  been made of the effects of toxic
substances on  the sexual behavior of laboratory animals. A  few
recent  references (50-61) are included in the listing  at the end of
this  chapter,  but they  deal  primarily with effects of drugs like
cannabis, alcohol, and morphine. A search of the literature between
 1978  and  1980  revealed  only two  references dealing  with other
agents, one  on  cyanogenic substances (62) and the other on lindane
(63). In addition, no systematic evaluations of sexual behavior have

-------
                               140

been conducted on humans known to have been exposed to toxic
substances either  in adulthood or  prenatally.  This unfortunate
circumstance,  that parallel studies have  not  been carried  out on
intentionally exposed animal subjects and on accidentally exposed
human  beings, severely  limits  the  capability to formulate either
qualitative or quantitative risk assessments for sexual functioning.
    Basic  research on human  sexual behavior should be strongly
encouraged at this time so  that appropriate demographic norms and
standards can be established. Adequate information on these matters
has not been developed  despite the pioneering efforts of Kinsey in
the late forties. In  addition,  changes in  concepts,  data gathering
techniques, and attitudes require modernization of the data base. As
pointed out in earlier sections of this discussion, neither measure-
ment of number of offspring produced, frequency of coitus, or even
frequency of  orgasm are adequate  as indicators of human sexual
functioning. There  is a strong need to develop epidemiological
studies of human sexual  behavior in its broadest scope and in terms
most meaningful to  human welfare and to the stability of interper-
sonal relationships.
    The scope of investigations of animal sexual behavior should  be
broadened.  Efforts to establish models  permitting better measure-
ment of sexual attractiveness, sexual motivation or desire, and even
sexual gratification should  be encouraged. Moreover, sound para-
metric data on the effects  of known environmental toxicants ought
to  be vigorously pursued. These  studies could be  carried out
profitably at this  time even with the limited  number of behavioral
measures currently available, and there should be strong  support for
such studies on a variety of species. The limitation of data bases,  no
matter how extensive, to rats and guinea pigs poses a serious obstacle
to  flexible choice of alternative models that  may in fact be more
appropriate to human problems.
    Finally, even  from the relatively  limited standpoint of sexual
behavior, more information is needed on how different classes of
chemical  substances  interact  with central neural  tissues  on the
cellular levels.  Information of this sort is fundamental not only to
interpretation of  toxicant effects on behavior,  but also to  sound
hypothesis formulation  and to development  of a framework that
would permit  prediction of the likely biological effects of a putative
toxicant.

-------
                                  141
                            REFERENCES

 1. Rausch, L., Klein, J., Shaffer, S. Q. and Stein, Z.: Effect of chlorination of
     drinking water on perinatal'mortality in New York villages,  1968-1977.
     Abstract. Am. J.Epidemiol. 112: 439, 1980.
 2. Levine, R. J., Symons, M. J., Balogh, S. A., Arndt, D. M., Kaswandik, N. 'T.
     and Gentile, J.W.: A method for monitoring the fertility of workers: I.
     Method and pilot studies. J. Occup. Med. 22: 781-791, 1980.
 3. Levine,  R. J., Symons, M. J., Balogh, S. A., Milby, T. H. and Whorton,
     M. D.: A method for monitoring the  fertility of workers. II. Validation of
     the method among workers exposed to dibromochloropropane. J. Occup.
     Med. 23: 183-188, 1981.
 4. Slone, D., Shapiro, S. and Miettinen, O. S.: Case-control surveillance of
     serious illnesses attributable to ambulatory drug use. In: Epidemiological
     Evaluation of Drugs, F.Colombo, S.Shapiro, D.Slone, and G.Tognoni,
     Eds.,  Elsevier North-Holland Biomedical Press:  Amsterdam; pp. 59—82,
     1977.
 5. Rosenberg, L., Shapiro, S., Slone, D., Kaufman, D. W., Miettinen, O. S.  and
     Stolley,  P.O.: Thiazides and  acute  cholecystits. N. Engl.  J. Med. 303:
     546-548, 1980.
 6. Mitchell,  A.   A.,   Rosenberg,  L.,  Shapiro,  S.  and Slone, D.:  Birth
     defects  related  to use  of Bendictine in pregnancy. I.  Oral clefts  and
     cardiac defects. J. Am. Med. Assoc. 245: 2311-2314, 1981.
 7. Selikoff, I. J., Churg, J. and Hammond, E. C.: Relation between exposure to
     asbestos and mesothelioma. New Engl. J. Med. 272: 560-565,  1965.
 8. Lee, A.  M. and Fraumeni, J. F., Jr.: Arsenic and  respiratory cancer in man:
     An occupational study. J. Natl. Cancer Inst. 42:  1045-1052, 1969.
 9. Harlap,  S,, Shiono, P., Ramcharan,  S., Berendes, H. and Pellegrin, F.: A
     prospective study of spontaneous fetal losses after induced abortions. New
     Engl. J. Med. 301: 677-681, 1979.
lO.Notari,  R. E.: Biopharmaceutics and Pharmacokinetics: An  Introduction.
     3rd Edition, Marcel Dekker: New York; 329 pp., 1980.
11. Wagner, J.  G.:  Biopharmaceutics and relevant  pharmacokinetics. Drug
     Intelligence Publications: Washington, D.C.; 375 pp., 1971.
12. Gibaldi, M. and Perrier, D.: Pharmacokinetics. Marcel Dekker:  New York;
     325 pp., 1975.
13. Bischoff, K. B.: Some fundamental considerations of the application of
     pharmacokinetics to cancer chemotherapy. Cancer Chemother. Rep. Part  1
     59: 777-796, 1975.
14. Himmelstein, K.  J.  and  Lutz,  R. J.: A review  of the applications of
     physiologically  based   pharmacokinetic  modeling.  J.  Pharmacokinet.
     Biopharm. 7: 127-145, 1979.
IS.Dedrick, R. L.: Animal scale-up. J. Pharmacokinet. Biopharm. 1: 435—461,
     1973.
16. Dedrick, R. L. and Bischoff, K.  B.: Species similarities in pharmacokinetics.
     Fed.Proc. 39: 54-59, 1980.

-------
                                   142

17. Gehring, P. J., Watanabe, P. G. and Blau, G.E.: Pharmacokinetic studies in
     evaluation of the toxicological and environmental hazard of chemicals: In:
     Advances  in  Modern Toxicology: New Concepts in Safety Evaluation,
     M. Mehlman, H. Blumenthal, and R. Shapiro, Eds., Hemisphere Publishing
     Corporation: New York; pp. 195-290, 1976.
18. World Health Organization. Chemobiokinetics and metabolism. In: Environ-
     mental  Health Criteria 6.  Principles and  Methods  for Evaluating the
     Toxicity of Chemicals. Geneva; pp. 116-177, 1978.
19.Filov, V. A.,  Golubev,  A.  A., Liublina, E.I. and Tolokontsev, N.A.:
     Quantitative Toxicology. John Wiley: New York; 462 pp., 1980.
20. Young, J. F.  and Holson, J. F.:  Utility of pharmacokinetics in  designing
     toxicological protocols and improving  interspecies extrapolation.  J.
     Environ. Pathol. Toxicol. 2: 169-186, 1978.
21.Souchay, A. M., Rice, P. C., Rourke,  I.E. and Nesbitt, R.E. L.: Glucose
     metabolism and transfer in the human placenta. In: Chemical Engineering
     in Medicine, D.D. Reneau, Ed., Plenum Press: New York; pp. 172-199,
     1973.
22. Dedrick,  R. L.,  Meyers, C.  E., Bungay, P. M. and De  Vita, V. T.:
     Pharmacokinetic   rationale  for  peritoneal drug administration  in  the
     treatment  of ovarian cancer. Cancer Treatment Rep. 62: 1—11, 1978.
23. Lee, I. P. and Dixon, R. L.:  Factors influencing reproduction and genetic
     toxic effects on male gonads. Environ.  Health Perspect. 24: 117—127,
     1978.
24. Beach, F. A.: Animal models  for human sexuality. In: CIBA Foundation
     Symposium, No.  62. Sex, Hormones, and Behavior, Mar. 14-16, 1978,
     London, England, Elsevier-North Holland: New York; pp.  133-144, 1979.
25.McClintock,  M. K. and Adler, N. T.:  The role  of the female during
     copulation in wild and domestic rats (Rattus norvegicus). Behavior 67:
     67-96, 1978.
26. Madlafousek, J. and Hlinak,  Z.: Sexual behavior of the female laboratory
     rat:  inventory,  patterning, and  measurement. Behavior 63: 129—174,
     1977.
27. Dewsbury, D. A.: A quantitative description of the behavior of rats during
     copulation. Behaviour 29: 154-178, 1967.
28. Dewsbury, D. A.: Description of  sexual  behavior in research  on hormone-
     behavior interactions. In: Endocrine Control of Sexual Behavior, C. Beyer,
     Ed., New York: Raven Press, pp. 3-32,1979.
29.McClintock, M. K.,  Toner, J. P., Adler, N. T. and Anisko, J. J.: Postejacu-
     latory  quiescence in  female and  male rats: consequences for sperm
     transport during group mating. J. Comp. Physiol. Psycho!.; in press, 1982.
30. Beach, F. A. and Jordan, L.: Sexual exhaustion and recovery in the male rat.
     Q. J. Exp. Psychol. 8: 121-133,1956.
Sl.Larsson, K.:  Conditioning and Sexual Behavior in the Male Albino Rat.
     Stockholm: Almquist & Wiksell;  1956.
32. Sachs, B. D. and Barfield, R.: Functional analysis of masculine copulatory
     behavior in the rat. Adv. Study Behav. 7: 91-154, 1976.

-------
 33

 34


 35

 36



 37


 38.


 39.

 40.


 41.


 42.



 43.


44


45.


46.

47.

48.

49.
                                 143

 .Young, W. C.: Psychobiology of sexual  behavior in  the guinea pig. Adv.
   Study Behav. 2: 1-110,1969.
 Young, W. C. and Grunt, J. A.: The pattern and measurement of sexual
   behavior in the male guinea pig. J. Comp. Physiol. Psychol. 44: 492-500
   1951.
 Dewsbury, D. A.: Copulatory behavior of rats: Variations within the dark
   phase of the diurnal cycle. Commun. Behav. Biol. 1: 373-377, 1968.
 Fadem, B.  H.,  Barfield,  R. J. and Whalen, R.E.: Dose-response  and
   time-response  relationships between progesterone  and the display of
   patterns of receptive and  preceptive behavior in the female rat. Horm.
   Behav. 13: 40-48,  1979.
 Goy, R. W. and Young, W. C.: Strain differences in the behavioral responses
   of female  guinea  pigs  to alpha-estradiol benzoate and  progesterone.
   Behaviour 10: 340-354, 1957.
 Walker, W. A. and Feder, H. H.: The comparative potency of various steroids
   to complete the priming process for lordosis in guinea pigs. Horm. Behav
   12: 299-308, 1979.
 Diakow, C. and Dewsbury, D. A.: A compuative description  of the mating
   behavior of female rodents. Anim. Behav. 26: 1091-1097,1978.
 McClintock, M. K., Anisko, J.J. and Adler, N.T.: Group mating among
   Norway rats.  I. Sex  differences' in the pattern  and  neuroendocrine
   consequences of copulation. Anim. Behav.; in press, 1982.
. Beach,  F. A.: Cross-species comparisons and the human heritage. In: Human
   Sexuality in Four Perspectives, F. A. Beach, Ed., Johns Hopkins University
   Press: Baltimore; pp., 296-316, 1977.
.Beach,  F. A.: Animal  models  and  psychological inference. In: Human
   Sexuality:   A  Developmental  and  Comparative   Approach,  H. A.
   Katchadourian, Ed., University of California Press: Berkeley ;pp  98-112
   1979.
. Adams, D., Gold, A. and Burt A.: Rise in female-initiated sexual activity at
   ovulation and its suppression by oral contraceptives. N Engl J Med 299-
   1145-1150,1978.
Persky,  H., Lief,  H., Strauss, D., Miller, W. and O'Brien, C.D.: Plasma
   testosterone  level and sexual behavior of couples. Arch. Sex. Behav. 7:
   157-173, 1978.
Byrne,  D.  and Scheffield, J.: Response to sexually arousing stimuli as a
   function of repressing and  sensitizing defenses. J. Abnorm. Psychol 70:
   114-118,1965.
Byrne,  D., Fisher, J. D., Lamberth, J. and Mitchell, H. E.: Evaluations of
   erotica: Facts or feelings. J. Pers. Soc. Psychol. 29: 111-116, 1974.
Kaats, A. and Davis,  K.:  Effects of volunteer biases in studies of sexual
   behavior and attitudes. J. Sex. Res. 7: 219-227, 1971.
Bauman, K.:  Volunteer bias in a study of sexual knowledge, attitudes, and
   behavior. J. Marr. Fam. 35: 27-31,1975.
Kaplan, H. S.: The New Sex Therapy, Brunner/Mazel: New York; 544 pp
   1974. '

-------
                                   144

50. Abel, E. L.: A review of alcohol's effects on sex and reproduction. Drug
     Alcohol Depend. 5: 321-332,1980.
51. Barnes, T. R., Bamber, R. W. and Watson, J.P.: Psychotropic drugs and
     sexual behaviour. Br. J. Hosp. Med. 21: 594-600,1979.
52. Clemens, L. G., Popham, T. V. and Ruppert, P. H.:  Neonatal treatment of
     hamsters with barbiturate alters adult sexual behavior.  Dev. Psychobiol.
     12: 49-59,1979.
SS.Dalterio,  S. L.: Perinatal or adult  exposure to cannabinoids  alters male
     reproductive functions  in  mice.  Pharmacol.  Biochem.  Behav.  12:
     143-153,1980.
54.Dalterio, S. and Bartke, A.: Perinatal exposure to cannabinoids alters male
     reproductive function in mice. Science 205: 1420-1422, 1979.
55. Gordon,  J.  H.,  Bromley,  B.  L.,  Gorski,  R.A.  and Zimmermann.E.:
     Delta9-tetrahydrocannabinol enhancement of lordosis behavior in estrogen
     treated female rats. Pharmacol. Biochem. Behav. 8: 603-608, 1978.
56. Kostellow, A. B., Ziegler, D., Kunar, J., Fujimoto, G. I. and Morrill, G. A.:
     Effect  of  cannabinoids  on estrous cycle,  ovulation,  and  reproductive
      capacity of female A/J mice. Pharmacology 21: 68-75, 1980.
57. Kumar, R., Mumford, L. and Teixeira, A. R.: Sexual behavior in morphine-
      dependent rats [proceedings]. Br. J. Pharmacol. 62: 389P-390P, 1978.
 58. Mumford, L. and Kumar, R.: Sexual behaviour of morphine-dependent and
      abstinent male  rats. Psychopharmacologie (Berlin) 65: 179-185, 1979.
 59. Ostrowski, N. L., Stapleton, J. M., Noble, R. G. and Reid, L. D.: Morphine
      and  naloxone's effects on sexual  behavior of the  female golden hamster.
      Pharmacol. Biochem. Behav. 11: 673-681, 1979.
 60. Rosenkrantz, H.: Effects on cannabis on fetal development of rodents. Adv.
      Biosci. 22-23: 479-499, 1978.
 61.Vorhees, C. V.,  Brunner, R. L.  and Butcher, R. E.: Psychotropic drugs as
      behavioral teratogens. Science 205: 1220-1225, 1979.
 62.Olusi, S. 0., Oke, O.  L. and Odusote, A.: Effects of cyanogenic agents on
      reproduction  and  neonatal development  in rats. Biol. Neonate  36:
      233-243,1979.
 63. Palmer, A. K., Cozens, D. D., Spicer, E. J. and Worden, A.N.: Effects of
      lindane upon  reproductive function in  a three-generation study in rats.
      Toxicology 10: 45-54,1978.

-------
                        STEERING COMMITTEE
K. Diane Courtney, Ph.D.
Research Pharmacologist
Pesticides and Toxic Substances
  Effects Laboratory
U.S. Environmental Protection
  Agency
Research Triangle Park, NC 27711

Wayne M. Galbraith, Ph.D.f
Toxicologist
Office of Research and Development
U.S. Environmental Protection
  Agency
Washington, D.C. 20460
(EPA Co-Project Officer)

Richard M. Hoar, Ph.D.
Head of Teratology and Assistant
  Director of Toxicology
Department of Toxicology
Hoffmann—LaRoche, Inc.
Nutley, NJ07110
(Chairman of Reproduction
  Groups)

E. Marshall Johnson, Ph.D.
Professor and Chairman
Department of Anatomy
Director, Daniel Baugh Institute
  of Anatomy
Jefferson Medical College
Thomas Jefferson University
Philadelphia, PA 19107
(Chairman of Developmental Group)
Robert M. Pratt, Ph.D.*
Chief, Experimental Teratogenesis
  Section
Laboratory of Reproductive &
  Developmental Toxicology
National Institute of Environmental
  Health Sciences
National Institutes of Health
Research Triangle Park, NC 27709
Michael G. Ryon, M.S.
Information Analyst
Chemical Effects Information
  Center
Oak Ridge National Laboratory
Oak Ridge, TN 37830
Peter Voytek, Ph.D.
Director, Reproductive Effects
  Assessment Group
Office of Research and
  Development
U.S. Environmental Protection
  Agency
Washington, D.C. 20460
(EPA Co-Project Officer)
 *Attended only the St. Louis workshop.
 •(•Currently, Acting Chief of the Toxicology Branch, Research Division, Chemi-
cal Systems Laboratory, Aberdeen Proving Ground, Maryland 21001.
                                   145

-------

-------
                             PARTICIPANTS
Rupert P. Amann, Ph.D.
Professor of Physiology and
  Biophysics
Animal Reproduction Laboratory
Colorado State University
Fort Collins, CO 80525

J. Michael Bedford, Ph.D.
Professor of Obstetrics and
  Gynecology and of Anatomy
Cornell Medical School
New York, NY 10021
(Chairman of Male Reproduction
  Group)

Allan R. Beaudoin,  Ph.D.
Assistant Chairman and Professor
Department of Anatomy
University of Michigan Medical
  School
Ann Arbor, MI 48104

Kenneth B. Bischoff, Ph.D.
Chairman, Department of Chemical
  Engineering
Professor of Biomedical and
  Chemical Engineering
University of Delaware
Newark, DEI 9711

William J. Bremner, M.D., Ph.D.
Chief, Endocrinology Section
Veterans Administration Medical
  Center
Associate Professor of Medicine
  and of Obstetrics and Gynecology
University of Washington School
  of Medicine
Seattle, WA 98108
Charles C. Brown, Ph.D.*
Statistician, Biometry Branch
National Cancer Institute
Bethesda, MD 20205

Mildred S. Christian, Ph.D.
Director of Research
Argus Research Laboratories
Perkasie, PA 18944

James H. Clark, Ph.D.
Professor of Cell Biology
Baylor College of Medicine
Houston, TX 77030
(Chairman of Female
  Reproduction Group)

Thomas F. X. Collins, Ph.D.
Chief, Mammalian Reproduction
  and Teratology
Division of Toxicology
Bureau of Foods
Food and Drug Administration
Washington, D.C. 20204
Donald A. Dewsbury, Ph.D.*
Professor of Psychology
University of Florida
Gainesville, FL 32611


Larry L. Ewing, Ph.D.*
Professor of Reproduction
  Biology
School of Hygiene and Public
  Health
Johns Hopkins University
Baltimore, MD 21205
  *Attended only the St. Louis workshop.
                                  147

-------
                                   148
Robert H. Foote, Ph.D.
Professor of Animal Science
Cornell University
Ithaca, NY 14850
David W. Gaylor, Ph.D.
Director, Division of Biometry
National Center for Toxicological
  Research
Jefferson, AR 72079
 Arnold A. Gerall, Ph.D.*
 Professor of Psychology
 Tulane University
 New Orleans, LA 70118
Robert W. Goy.Ph.D.*
Director, Wisconsin Regional
  Primate Research Center
University of Wisconsin
Madison, WI 53706
(Chairman of Behavior Group)
 Arthur F. Haney, M.D.
 Assistant Professor of Obstetrics
  and Gynecology
 Duke University Medical Center
 Durham, NC 27710
 W. LeRoy Heinrichs, Ph.D., M.D.*
 Chairman and Professor
 Department of Gynecology and
   Obstetrics
 Stanford University
 Stanford, CA 94305
Mary C. Henry, Ph.D.*
Research Pharmacologist
Environmental Protection Research
  Division
U.S. Army Biomedical Engineering
  Research and Development
  Laboratory
Fort Detrick
Frederick, MD 21701

Jerry Highfffl, M.S.
Statistician, Health Effects
  Research Laboratory
U.S. Environmental Protection
  Agency
Research Triangle Park, NC 27711

Carol J. Hogue, Ph.D.
Associate Professor of Biometry
University of Arkansas for
  Medical Sciences
Little Rock, AR 72201

Donald E. Hutchings, Ph.D.
Research Scientist
Department of Behavioral
  Physiology
New York State Psychiatric
  Institute
Assistant Professor of Medical
  Psychology
Department of Pediatrics
Columbia University
New York, NY 10031

Harold Kalter, Ph.D.
Research Associate
Children's Hospital Research
  Foundation
Professor of Research
Department of Pediatrics
University of Cincinnati
Cincinnati, OH 45229
  *Attended only the St. Louis workshop.

-------
                                    149
Carole A. Kimmel, Ph.D.*
Chief, Perinatal and Postnatal
  Evaluation Branch
Research Pharmacologist
Division of Teratogenesis
  Research
National Center for Toxicological
  Research
Jefferson, AR 72079

Devendra M. Kochhar, Ph.D.
Professor of Anatomy
Jefferson Medical College
Thomas Jefferson University
Philadelphia, PA 19107

Donna Kuroda, Ph.D.
Physical Sciences Administrator
Reproductive Effects
  Assessment Group
U.S. Environmental Protection
  Agency
Washington, D.C. 20460

Donald R. Mattison, M.D.
Medical Officer
Pregnancy Research Branch
National Institute of Child
  Health and Human Development
National Institutes of Health
Bethesda, MD 20205

Martha K. McClintock, Ph.D.*
Assistant Professor of Behavioral
  Sciences
University of Chicago
Chicago, IL 60637

Wilbur P. McNulty, M.D.
Chairman, Laboratory of Pathology
Oregon Regional Primate Research
  Center
Beaverton, OR 97006
 Marvin L. Meistrich, Ph.D.
 Associate Professor of
  Experimental Radiotherapy
 University of Texas System
  Cancer Center
 M.D. Anderson Hospital Tumor
  Institute
 Houston,  TX 77030

 Eugene F. Oakberg, Ph.D.*
 Senior Research Staff Member
 Mammalian Genetics and
  Development Section
 Biology Division
 Oak Ridge National Laboratory
 Oak Ridge, TN 37830

 James W.  Overstreet, Ph.D., M.D.*
 Associate  Professor of Human
  Anatomy and of Obstetrics and
  Gynecology
 University of California, Davis
  Medical School
 Davis, CA 95616

John C. Porter, Ph.D.
Professor of Physiology and of
  Obstetrics and Gynecology
University of Texas Health Science
  Center at Dallas
Southwestern Medical School
Dallas, TX 75235

Lynn Rosenberg, Sc.D.
Assistant Research Professor
Boston University
Biostatistician and
  Epidemiologist
Drug Epidemiology Unit
Boston University Medical
  Center
Cambridge, MA 02138
  *Attended only the St. Louis workshop.

-------
                                   150
Griff T. Ross, Ph.D.,M.D.f
Deputy Director of the
  Clinical Center
National Institutes of Health
Bethesda, MD 20205

Liane B. Russell, Ph.D.f
Head of Mammalian Genetics and
  Teratology Section
Biology Division
Oak Ridge National Laboratory
Oak Ridge, TN 37830

Carol Sakai, Ph.D.
Reproductive lexicologist
Reproductive Effects Assessment
  Group
Office of Research and
  Development
U.S. Environmental Protection
  Agency
Washington, D.C. 20460

Thomas H. Shepard, M.D.*
Professor of Pediatrics
Head, Central Laboratory for
  Human Embryology
University of Washington
Seattle, WA 98195

Richard G. Skalko, Ph.D.
Professor and Chairman
Department of Anatomy
College of Medicine
East Tennessee State University
Johnson City, TN 37614
Kate Smith, Ph.D.*
Developmental Toxicologist
Health Effects Research Laboratory
U.S. Environmental Protection
  Agency
Cincinnati, OH 45268

Robert E. Staples, Ph.D.
Staff Teratologist
Haskell Laboratory (DuPont)
Wilmington, DE 19898


Robert G. Tardiff, Ph.D.f
Executive Director, Board on
  Toxicology and Environmental
  Health Hazards
National Academy of Sciences
Washington, D.C. 20418
William J. Waddell, M.D.
Professor and Chairman
Department of Pharmacology and
  Toxicology
School of Medicine
University of Louisville
Louisville, KY 40292


Ronald J. Young, Ph.D.
Associate Professor and Research
  Associate
Department of Obstetrics and
  Gynecology
Cornell University Medical College
New York, NY 10021
  * Attended only the St. Louis workshop.
  t Attended only the Atlanta workshop.

-------
                              REVIEWERS

  The  assistance of the following  persons in the review  process is gratefully
acknowledged. The final content of the report is the responsibility of the steer-
ing committee and the group chairmen.
Aaron Blair, Ph.D.
Environmental Epidemiology
  Branch
National Institutes of Health

Joseph Borzelleca, Ph.D.
Department of Pharmacology
Medical College of Virginia

Robert Dedrick, Ph.D.
Biomedical Engineering and
  Instrumentation Branch
National Institutes of Health

James Emerson, Ph.D.
Life Sciences
Coca Cola Company

Michael Farrow, Ph.D.
Genetic Toxicology Department
Hazleton Laboratories America,
  Inc.

Ernst Freese, Ph.D.
Laboratory of Molecular Biology
National Institutes of Health

Vera Glocklin, Ph.D.
Bureau of Drugs
U.S. Food and Drug Administration

Andrew G. Hendrickx, Ph.D.
California Primate Research
  Center
University of California, Davis

Kundan S. Khera, Ph.D.
Health Protection Branch
Health and Welfare Canada
Renate Kimbrough, M.D.
Toxicology Branch
Centers for Disease Control

George Levinskas, Ph.D.
Environmental Assessment and
  Toxicology
Monsanto Company

Lawrence B. Mellett, Ph.D.
Scientific Liaison and Compliance
Revlon Health Care Group

Roy Mundy, Ph.D.
Department of Pharmacology
University of Alabama,
  Birmingham

Frederick Oehme, D.V.M., Ph.D.
Comparative Toxicology Laboratory
Kansas State University

Anthony K. Palmer
Huntingdon Research Centre

Bobby Joe Payne, D.V.M., Ph.D.
Director of Pathology
Toxicity Research Laboratories,
  Ltd.

Harold M. Peck, M.D.
Safety Assessment
Merck Institute for Therapeutic
  Research

David Rail, M.D., Ph.D.
National Institute of Environmental
  and Health Sciences
                                   151

-------
                                   152
Robert Scala, Ph.D.
Medical and Environmental Health
  Department
Exxon Corporation

Charlotte Schneyer, Ph.D.
Laboratory of Exocrine Physiology
University of Alabama,
  Birmingham

Bernard A. Schwetz, D.V.M., Ph.D.
Health and Environmental
  Sciences
Dow Chemical U.S.A.
Marshall Steinberg, Ph.D.
Life Sciences Division
Hazleton Laboratories America, Inc.

Clarence J. Terhaar, Ph.D.
Toxicology Section
Eastman Kodak Company

Hanspeter Witschi, M.D.
Biology Division
Oak Ridge National Laboratory

Gerhard Zbindin, M.D.
Institute of Toxicology

-------
                                  INDEX
 abstinence interval, 60, 82
 acceptable daily intake (ADI), 9,
  48,56, 110
 acceptable dosage level, 48, 111,
  112
 accessory sex glands, 6,47 (Table 8),
  49 (Table 9), 50 (Table  10), 51,
  60,73,75-77,79,84
 acute, 3,57, 70, 71,99, 122
 adenosis, 8 (Table 1), 37
 adolescence, 126
 adrenal, 8 (Table 1), 127
 adult toxic dose, 108,  111,112
 age, 7, 10,11, 14 (Table 2), 16,
  19-21, 28, 48 (Table 8), 50
  (Table 10), 52, 53, 60, 74, 75,
  82, 111, 118
 amenorrhea, 20, 37, 39
 androgen, 5, 6,14, 19, 20, 30, 33
  (Table 6), 37,  62, 73, 93,126,
  128-130
 animal
  model, 11, 17, 18,20,23,28,
    30,32, 41-43,44 (Table 7),
    50 (Table 10), 52, 56, 57, 58
    (Table 11), 60, 62, 69, 71,83,
    110 (Table 12), 122, 124,
    128-130, 140
  testing, 2, 6, 13, 19, 22,25-27,
    42, 46, 47 (Table 8), 49
    (Table 9), 50 (Table 10), 52,
    55-57, 58 (Table 11), 59-61,
    81,85,86,100,101,  104,106,
    108, 109,  110(Table  12), 111,
    112,119,  130-137, 139, 140
anovulation, 8 (Table 1), 17, 20, 37
 artificial insemination, 43, 44
  (Table 7), 58 (Table  11), 85
 attractivity, 124, 125, 131, 134,
   135,140
 azoospermia, 54, 59
 bioaccumulation, 48, 57, 58
  (Table 11), 59, 102, 108
 breast, 8 (Table 1)
case-control study, 118-120
cauda epididymidis, 44 (Table 7),
  49,51,59,72,74,83
cell culture, 22-24, 31, 32, 33
  (Table 6), 62, 109, 110
  (Table 12)
chronic, 8 (Table 1), 46, 48
  (TableS), 51,70, 71,88,99,
  111,  122
cleft lip, 101
cleft palate, 101
coefficient of variation (CV), 46,
  50 (Table 10), 60, 62, 75, 77, 81
cohort-studies, 118, 119, 121, 137
computerized integrated data base,
  4 (Fig. 1), 5-7, 9, 10, 121
conception, 43, 118
conceptive ability, 6, 17, 31
conceptus, 1, 99, 100, 103, 104,
  106-110, 112, 122
congenital defects (or malforma-
  tions), 100, 118
control groups, 16, 46, 53-57, 61,
  70-72, 74, 77, 78, 80-84, 123,
  132, 137-139
copulation plug, 16
copulatory behavior, 17,132-134
                                   153

-------
corpus luteum (lutea), 17,30, 32,
  37,47 (Table 8), 50 (Table 10),
  83,85
correlational studies, 118
cost/benefit, 16, 17, 49 (Table 9),
  108,139
critical periods of development,
  20,102, 103
decidual implantation, 104, 105
demographic studies, 3, 118
developmental abnormalities (or
  defects or malformations), 99,
  100, 103-107,110 (Table 12),
  118,126,129,130
developmental toxic dose, 108,
  110(Table 12), 111, 112
diestrus, 20,37
DNA, 31, 49 (Table 9), 62, 87
dog, 43, 44 (Table 7), 79, 131
domestic animals, 32, 42, 43,46,
  70, 74, 76, 79, 107, 108
dopamine, 11, 21, 24,37
dose, 6, 16, 18-20,22,26,27,
  47 (Table 8), 51,52, 55-57,
  59,61,83,87,88,95,101-104,
  106-109,111,121,122, 130,
  134, 136
dose-response (relationship), 2, 9,
  18, 20, 23, 24, 26-28,47 (Table 8),
  48, 57, 79, 85, 87, 88,101,103,
  109-111, 112 (Table 12), 119
ductus deferens, 72
ejaculate, 41-43, 46,47 (Table 10),
  49, 50 (Table 10), 51, 54, 55, 59,
  60, 69, 73-79, 82, 84, 85, 87, 94,
  133
ejaculation, 42, 43, 44 (Table 7),
  51,60,74,76,77,94, 132,133
emoryo, 47 (Table 8), 50 (Table 10),
  59,83,85,86, 101-103,105,
  109, 110 (Table 12), 112
154
  embryonic development, 41, 62,
   102-105
  emission, 95
  endocrine, 1, 26,43, 46,47
   (Table 8), 50 (Table 10), 53, 79,
   80, 103, 105,  107, 126, 134
  epidemiology (epidemiologic), 1,
   3,5,12,53,56, 106,117,119-
   121, 136-138, 140
  epididymis, 41, 46,47 (Table 8),
   49 (Table 9), 50 (Table 10), 51,
   59, 70, 72-75, 78, 83, 84
  estradiol, 14, 18, 19, 22, 26, 33
   (Table 6), 37, 127, 129, 134,
   136
  estrogen, 5, 9, 11, 13, 14 (Table 2),
   18,19,21,23,25-27,30,33
   (Table 6), 37, 38, 126-129, 136,
   138
  estrogenicity, 5, 6, 9, 11, 13,  14,
   18, 19, 21-23, 27
  estrus, 14, 15 (Table 3), 16, 17,
   20, 38, 104, 133, 134, 136
  extrapolation, 10-12,18-20, 30,
   32,56, 100, 102, 104, 111, 112,
   117,122, 123, 130, 136
 fallopian tubes, 8 (Table 1)
 false negative, 16, 17, 109
 false positive, 5, 17, 109
 fecundity, 17
 fertility,  16, 17, 28,4143,44
   (Table 7), 46,47 (Table 8), 50
   (Table  10), 51,53-56, 59, 60,
   78,83-87, 107,118,120
 fertilization, 41,43, 62
 fetal development, 10, 14, 20, 62,
   102-105, 129
 fetus, 2, 5, 102, 105, 106, 122,
   129, 130
 follicle, 11,21, 28, 29 (Table 4),
   30, 32, 33 (Table 6), 38,135
 follicle-stimulating hormone (FSH),
   21, 30, 38, 41,47 (Table 8), 50
   (Table  10), 53,54, 59,61,62,
   79-84, 95

-------
 gamete, 8 (Table 1), 27, 28, 104
 germ cell, 49 (Table 9), 71, 74
 germinal epithelium, 49, 54, 57,
  59,69,71,73,78
 gestation, 8 (Table 1), 18, 20,
  102-104
 gonad, 30, 54, 104, 122,  124, 126,
  128, 136
 gonadotropin, 10, 17, 21, 22, 26,
  30-32, 38, 136
 gonadotropin-releasing hormone
  (GnRH), 21, 22, 24, 47 (Table 8),
  50 (Table 10), 54, 61, 80, 82
 granulosa, 30, 32, 33 (Table 6)
 gross anatomical (or structural)
  defects, 106,107
 guinea pig, 17, 27, 127-129, 131,
  133-135, 140
hamster, 43, 47 (Table 8), 82, 84,
  86, 87, 127, 129
hazard, 1,6,41-43, 53, 62, 69, 85,
  99, 100, 106, 108, 109, 111,
  112,  117-121
heterospermic insemination, 62
hormone, 5, 6, 8 (Table 1), 9,11,
  21,25,27, 30, 33 (Table 6),
  41, 44 (Table 7), 47 (Table 8),
  50 (Table 10), 53,54, 59,61,
  62,79-84,95,124,126-131,
  133,  136
human, 1-3,  5-7, 9-12,  16-20, 22,
  24, 27, 28, 30-32, 41-43, 44
  (Table 7), 47 (Table 8), 49
  (Table 9), 50 (Table 10), 52-62,
  70, 75, 76, 79-82, 84-88, 99-
  102,  106-112, 117, 119,120,
  122-130, 134-140
human chorionic gonadotropin
  (HCG),31
hypothalamus (hypothalamic), 5,
  8 (Table 1), 11, 20,21,24-27,
  38,80, 128, 131
hypothesis-generating studies,
  117-119
155
  implantation, 14, 104
  infertility, 1, 5, 42, 43, 52, 54, 55,
   80,82,118
  intromission, 132, 133
  inutero,  104, 106
  in vitro, 10,  18, 22-24, 30, 32, 33
   (Table 6), 44 (Table 7), 47
   (Table 8), 61,62, 79,82,84,
   85, 109, 110 (Table 12)
  in vivo, 10, 14, 18,19, 21-23, 30,
   31,57,62,79,85,86,108, 109
 lactation, 6, 8 (Table 1), 14, 16,
   18,38,104,105
 leydigcell, 61,62, 72
 libido, 20,43, 84, 124,127, 132,
   140
 litter size, 18, 84
 live birth index, 18
 lordosis, 26, 27, 135
 luteinizing hormone (LH), 21, 22,
   25, 30, 38, 41,47 (Table 8),
   50 (Table 10), 54,59,61,62,
   79-84, 95
 masculinization, 5,10, 20,40,
   126, 129
 maternal-fetal exchange, 102
 maternal toxicity, 110
 mating, 14,  15 (Table 3), 16, 17,
   26, 27, 44 (Table 7), 46, 58
   (Table 11), 59, 62,83, 85,86,
   104, 133
 mating behavior, 14, 20, 25, 26
 maximum tolerated dose (MTD),
   16,47 (Table 8), 49 (Table 9),
   57, 83, 88, 95
 menopause, 8 (Table 1), 28, 38
 menstruation, 17; 30, 138
 model system, 9-11, 28, 32, 43,
   60,69,71, 101, 110 (Table 12),
   122, 124,  130
 motility (or motile), 42,43, 46,
   47 (Table  8), 50 (Table 10), 51,
   53, 60, 72, 73, 75, 77-79, 82-84,
   95

-------
mounts, 124, 126, 132, 133, 135
mouse (mice), 6, 11, 28, 43,44
  (Table?), 51,61,74, 102,  110
  (Table 12), 127,129
multigenerational, 6, 15 (Table 3),
  104, 105
nonmotile spermatozoan, 95
no-observed-effect level (NOEL)
  or (no-adverse-effect level), 56,
  103, 110
oligomenorrhea, 20, 38
oligozoospermia, 54
oocyte, 8 (Table 1), 11, 18,27,
  28, 29 (Table 4), 39, 84
oogenesis, 28
oral contraceptive, 11
organogenesis, 101, 102, 105,
  106,122
ovary, 8 (Table 1), 16-18, 21, 25,
  27, 28, 30-32, 33 (Table 6),
  127, 134-136
ovotoxicity, 11
ovulation, 10,  14, 17,20, 21, 39
parturition, 8 (Table 1), 39
pharmacokinetics, 1,56, 101,
  103, 111, 112, 117, 121-123,
  130
phenotypic transformation, 20
pituitary, 8 (Table 1), 10, 21-26,
  54, 80, 128
placenta, 8 (Table 1), 102, 103,
  112, 129,130
placental transfer, 8 (Table 1), 102
population-based registries, 118
postpartum, 14, 15 (Table 3), 16
potency, 18, 19,22, 23, 128
pregnancy, 5, 6, 8 (Table 1), 16,
  20,47 (Table 8), 50 (Table 10),
  52,105,110,117,118, 120
156
 primate, 10, 17, 22, 23, 30,43,
   44 (Table 7), 74, 131
 proceptivity, 135
 progestagen, 26, 136
 progesterone, 11,21, 26, 27, 30,
   33 (Table 6), 39, 136
 prolactin, 8 (Table 1), 21, 23, 24,
   39
 prostaglandin, 39
 puberty, 6, 8 (Table 1),  14, 39, 75,
   49 (Table 9)
 qualitative, 22, 43, 74, 87
   reproductive toxicity screen, 3,
    4 (Fig. 1), 6, 7,9, 13, 18, 19
   risk assessment, 46, 100, 128,
    140
   test, 10, 71
 Quantitative, 21, 25, 28, 43, 74,
    87, 121, 128
   reproductive toxicity test, 3,4
    (Fig. 1), 7, 10, 18, 24, 55, 71
   risk, 4, 5,32,46, 56,108, 110,
    140
 rabbit, 43,44 (Table 7), 46,47
  (Table 8), 49 (Table 9), 50
  (Table 10), 51, 56, 57, 58
  (Table 11), 59, 60,62, 70, 74,
  76,77,79,81,83,85,102,105
 rat, 13, 14 (Table 2), 17,19, 20,
  22, 23, 25, 26, 32, 43, 44
  (Table 7), 47 (Table 8), 49
  (Table 9), 50 (Table 10), 51,
  56, 57, 58 (Table 11), 59-62,
  73, 74, 79, 81, 83, 85, 102,
  105, 110 (Table  12), 124, 127-
  129, 131, 133-135, 140
 receptivity, 9, 124, 126, 127,132,
  134-136
 registries of birth defects, 120
 radioimmunoassay, 22,23, 31,
  81,82, 130

-------
 reproductive capabilities, 6, 8
   (Table 1)
 reproductive capacity, 62
 reproductive dysfunction, 49
   (Table 9), 53, 55, 82
 reproductive function, 20, 41,
   42, 53, 56-58, 137
 reproductive performance, 14, 55
 reproductive toxicants, 3, 6, 7, 9-
   11,48,52,54,80,81,86,123,
   137
 research, 2, 9-12, 49 (Table 9),
   60-62,76, 112, 120, 121, 123,
   139, 140
 reversibility, 2, 7,26,47 (Table 8),
   59,81,126, 139
 risk assessment, 1, 3, 4 (Fig. 1), 7,
   11-13,42,52,56,57,61,69,
   86,99, 117, 121,  123,139
 risk estimation, 2, 99,  100, 109,
   111,112
 rodent, 17, 18, 20, 27
 route of administration (or
   exposure), 9, 16, 26,27, 57, 83,
   101, 121, 130
 safety factor, 9, 52, 56, 57, 110-
  112
 screening system (or procedures),
  3, 4 (Fig. 1), 5-7, 14 (Table 2),
  16-18, 25, 27, 46, 47 (Table 8),
  50 (Table 10), 53,54, 57, 61,
  62,71,74,78,79,87,107,109,
  110 (Table 12), 124, 129, 131,
  137
scrotol circumference, 69, 70
semen, 41-43, 44 (Table 7), 46,
  47 (Table 8), 49, 50 (Table 10),
  51, 53-55, 58 (Table  11), 59-62,
  69, 73-79,82, 85-87, 96
seminal
  characteristics, 60, 70, 74, 75
  collections, 46,  74, 76, 82
157
   fluid (or plasma), 44 (Table 7),
     47 (Table 8), 49 (Table 9), 50
     (Table 10), 53,54,59-61,75,
     76, 79, 80, 82
   volume, 47 (Table 8), 50 (Table 10),
     60, 74, 75, 82, 84, 96
 seminiferous epithelium, 44 (Table 7),
   47 (Table 8), 48, 51,57, 71,74,
   75,83,84,88,93,94,96
 Sertoli cell, 62, 71,72,83,84
 sexual
   behavior, 1, 8 (Table 1), 10, 20,
     25-27,43,62,80,117,123-131,
     134-140
   dysfunction, 53, 123, 125., 137-139
   function, 26, 80, 104,124,137,
     138, 140
   gratification, 125, 138-140
   initative, 124-126, 131,138
   responsiveness,  124-126, 131,
     135,136
 short-term test, 31,62, 107-109,
   110 (Table 12)
 sonography, 53
 species, 7, 9,  11, 16, 17, 28, 30-32,
   43, 46, 49 (Table 9), 51,56,
   57,61,69-71,74-76,79-81,
   100-106, 110-112, 117, 122-124,
   126, 128-131, 134, 140
 sperm, 16, 41-43,  44 (Table 7),
    46, 47 (Table  8), 49 (Table 9),
    50 (Table 10), 51,54, 59, 69,
    62,69, 71, 72, 74-76, 78-80,
    82, 84-87, 132
  abnormalities, 60-62, 78, 85, 87
  chromatin, 62
  concentration, 41, 42, 46, 47
    (Table 8), 50 (Table 10), 72,
    76, 77, 82
  count, 42, 53, 54, 72, 76, 138
  genone, 41,62,85, 86
  head, 55, 62, 73, 78, 83
  maturation, 43
  morphology, 41, 42, 46, 47
    (Table 8), 50 (Table 10), 51,
    53, 55, 59-62,  73, 75, 78, 79,
    84,87

-------
motility, 42,43,46,47 (Table 8),
  50 (Table 10), 51,53,60,72,
  73, 75, 77-79, 82-84, 93, 95
number, 41,46,47 (Table 8), 50
  (Table 10), 53, 55,72, 76-79,
  82, 84, 85, 98
production, 42,44 (Table 7), 51,
  54, 60, 70, 75-77, 80, 85, 94 ,
size, 55
transport, 44 (Table 7), 47
  (Table 8), 59, 62, 70, 85
spermatid, 44 (Table 7), 47
  (Table 8), 50 (Table 10), 51,
  58,70,71,83,84
spermatocytes, 44 (Table 7), 47
  (Table 8), 49, 51,71,72, 83, 84
spermatogenesis, 43, 70, 71, 74,
  78, 80, 86, 94, 97
spermatogonia, 42,44 (Table 7),
  49,51,59,71
spermatozoa, 42,46,49 (Table 9),
  55, 60, 62, 69, 70, 72-74, 78,
  83, 85-87,94-98
spermatozoal concentration, 42,
  76,84
spermicidal, 79
spermiogenesis, 51
spontaneous luteinization, 32
sterility, 71, 80,82,86
steroid, 16,17,21,28,30,31
  (Table 5), 32, 33 (Table 6),
  •127,136,137
steroidogenesis, 8 (Table 1), 10,
  16,21,28-30, 31 (Table 5), 32,
  33 (Table 6), 39,136,137
structure-function, 3-7, 9, 10, 56 .
subchronic, 3,48, 57
surveillance study, 52, 54,120,
   121, 136, 137
158
  testis, 42,44 (Table 7), 47
    (Table 8), 49 (Table 9), 50
    (Table 10), 51,54, 56, 69,70,
    71,73,74,79,80,83-85, 127,
    128, 131
  testis weight, 44 (Table 7), 47
    (TableS), 50 (Table 10), 58
    (Table 11), 70, 77, 83, 84
  testosterone (T), 6, 20, 30, 33
    (Table 6), 39, 41,47 (Table 8),
    48, 50 (Table 10), 54, 59, 61,
    62,69,79-84,97,128, 129,131
  thecol, 30, 32, 33 (Table 6)
  threshold level, 2, 42, 56, 88, 103,
    110, 111, 130
  thyrotropin, 23
  tonometry, 49 (Table 9), 53, 60
  uncertainty factor, 56
  uterus (or uterine), 5, 8 (Table 1),
    13, 14 (Table 2), 18, 19, 112,
    122
   vagina, 8 (Table 1), 43, 59, 74, 84,
     132,  133, 136, 138
   vaginal
     opening, 5, 13,14 (Table 2), 18,
      20
     smear, 16, 20, 58 (Table 11),
      135
   validation, 7, 10, 16, 32, 46, 49
     (Table 9), 55, 84, 85, 99,  108,
     109,112,119, 120
   vesicular gland, 47 (Table 8), 50
     (Table 10), 73, 78, 83
   videotape (video),  46,47 (Table 8),
     48, 50 (Table 10), 60, 73,  78, 82
   virilization, 8 (Table 1), 40,  126
 testicular
  function, 41,42, 46, 52-56,58,
    60,61,69,71,72,74,78-80,
    82
  size, 44 (Table 7), 47 (Table 8),
    50 (Table 10), 51,53, 54, 60,
    69, 70, 74,84
   zona pellucida, 84, 86
   zygote, 8 (Table 1), 104
         US. GOVERNMENT PRINTING OFBCE: 1984 - 759-102/10704

-------

-------

-------